Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Bone ; 184: 117086, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38552893

ABSTRACT

PURPOSE: Mitofusin 2 (Mfn2) is one of two mitofusins involved in regulating mitochondrial size, shape and function, including mitophagy, an important cellular mechanism to limit oxidative stress. Reduced expression of Mfn2 has been associated with impaired osteoblast differentiation and function and a reduction in the number of viable osteocytes in bone. We hypothesized that the genetic absence of Mfn2 in these cells would increase their susceptibility to aging-associated metabolic stress, leading to a progressive impairment in skeletal homeostasis over time. METHODS: Mfn2 was selectively deleted in vivo at three different stages of osteoblast lineage commitment by crossing mice in which the Mfn2 gene was floxed with transgenic mice expressing Cre under the control of the promoter for Osterix (OSX), collagen1a1, or DMP1 (Dentin Matrix Acidic Phosphoprotein 1). RESULTS: Mice in which Mfn2 was deleted using DMP1-cre demonstrated a progressive and dramatic decline in bone mineral density (BMD) beginning at 10 weeks of age (n = 5 for each sex and each genotype from age 10 to 20 weeks). By 15 weeks, there was evidence for a functional decline in muscle performance as assessed using a rotarod apparatus (n = 3; 2 males/ 1 female for each genotype), accompanied by a decline in lean body mass. A marked reduction in trabecular bone mass was evident on bone histomorphometry, and biomechanical testing at 25 weeks (k/o: 2 male/1 female, control 2 male/2 female) revealed severely impaired femur strength. Extensive regional myofiber atrophy and degeneration was observed on skeletal muscle histology. Electron microscopy showed progressive disruption of cellular architecture, with disorganized sarcomeres and a bloated mitochondrial reticulum. There was also evidence of neurodegeneration within the ventral horn and roots of the lumbar spinal cord, which was accompanied by myelin loss and myofiber atrophy. Deletion of Mfn2 using OSX-cre or Col1a1-cre did not result in a musculoskeletal phenotype. Where possible, male and female animals were analyzed separately, but small numbers of animals in each group limited statistical power. For other outcomes, where sex was not considered, small sample sizes might still limit the strength of the observation. CONCLUSION: Despite known functional overlap of Mfn1 and Mfn2 in some tissues, and their co-expression in bone, muscle and spinal cord, deletion of Mfn2 using the 8 kB DMP1 promoter uncovered an important non-redundant role for Mfn2 in maintaining the neuromuscular/bone axis.


Subject(s)
Bone Density , GTP Phosphohydrolases , Animals , Female , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Male , Mice , Bone Density/genetics , Bone Density/physiology , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Bone and Bones/pathology , Bone and Bones/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Osteoblasts/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics
2.
Bone ; 145: 115835, 2021 04.
Article in English | MEDLINE | ID: mdl-33360005

ABSTRACT

In 2003, we briefly reported the remarkable osteopathy of a 12-year-old boy who at age two months began fracturing his limbs with subsequent hyperplastic callus formation and expansion and fusion of appendicular bones. By age ten years he had coalesced his lumbosacral spine, pelvis, femurs, and leg and foot bones as a single structure. Computed tomography of expanded bone revealed a thin cortical shell, diminished irregular trabeculae, and cystic areas. Histopathology featured foci of woven bone, densely packed osteocytes, cartilage, fibrovascular tissue, and massive fat deposition in the marrow space lacking hematogenous precursor cells. Bone turnover markers indicated accelerated remodeling and the few radiographically assessable appendicular bones improved during brief adherence to alendronate therapy. Following puberty, serum multiplex biomarker profiling confirmed accelerated bone turnover. At age 23 years, macrospecimens from leg amputation revealed ossification along capsular tissue together with hyaline cartilage degeneration. Concurrently, the life-long course of this same disorder was delineated in an unrelated woman until her death at age 51 years. Both patients demonstrated the radiographic hallmarks and harbored the heterozygous point mutation (c.-14C>T) in the 5'-UTR of IFITM5 associated with osteogenesis imperfecta type V (OI-V). Herein, we detail the clinical, radiological, histopathological, biochemical, and molecular findings and discuss the etiology and pathogenesis of this extraordinary osteopathy that we call coalescing expansile skeletal disease.


Subject(s)
Osteogenesis Imperfecta , 5' Untranslated Regions , Adult , Bone and Bones , Child , Female , Humans , Infant , Male , Membrane Proteins/genetics , Middle Aged , Mutation/genetics , Osteogenesis Imperfecta/diagnostic imaging , Osteogenesis Imperfecta/genetics , Young Adult
3.
Bone ; 131: 115137, 2020 02.
Article in English | MEDLINE | ID: mdl-31756522

ABSTRACT

Autosomal dominant hypophosphatemic rickets (ADHR) is remarkable among the hypophosphatemic rickets syndromes for its variable age of presentation and periods of quiescence during which serum phosphate and fibroblast growth factor 23 (FGF 23) levels are normal without therapy. In contrast, hypophosphatemia in X-linked hypophosphatemic rickets (XLH) manifests soon after birth and requires lifelong therapy. This suggests that there are environmental factors which can alter FGF 23 activity in ADHR but not in XLH. We present an adult with ADHR in whom resolution of hypophosphatemia was achieved by correcting iron deficiency without the need for phosphate supplementation. Serial iron and FGF 23 levels revealed an inverse relationship (r=-0.79, p<0.04). All patients with ADHR who present with hypophosphatemia and worsening symptoms should be screened for iron deficiency. If iron deficiency is detected, therapy with a combination of calcitriol and iron supplementation should be considered without phosphate supplementation.


Subject(s)
Familial Hypophosphatemic Rickets , Hypophosphatemia , Adult , Calcitriol , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/drug therapy , Familial Hypophosphatemic Rickets/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Humans , Iron , Phosphates
4.
Nat Commun ; 9(1): 2193, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29875355

ABSTRACT

Atherosclerosis and osteoporosis are epidemiologically linked and oxidation specific epitopes (OSEs), such as phosphocholine (PC) of oxidized phospholipids (PC-OxPL) and malondialdehyde (MDA), are pathogenic in both. The proatherogenic effects of OSEs are opposed by innate immune antibodies. Here we show that high-fat diet (HFD)-induced bone loss is attenuated in mice expressing a single chain variable region fragment of the IgM E06 (E06-scFv) that neutralizes PC-OxPL, by increasing osteoblast number and stimulating bone formation. Similarly, HFD-induced bone loss is attenuated in mice expressing IK17-scFv, which neutralizes MDA. Notably, E06-scFv also increases bone mass in mice fed a normal diet. Moreover, the levels of anti-PC IgM decrease in aged mice. We conclude that OSEs, whether produced chronically or increased by HFD, restrain bone formation, and that diminished defense against OSEs may contribute to age-related bone loss. Anti-OSEs, therefore, may represent a novel therapeutic approach against osteoporosis and atherosclerosis simultaneously.


Subject(s)
Epitopes/immunology , Immunoglobulin M/immunology , Osteogenesis/immunology , Osteoporosis/immunology , Single-Chain Antibodies/immunology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Diet, High-Fat/adverse effects , Epitopes/genetics , Epitopes/metabolism , Immunoglobulin M/genetics , Lipoproteins, LDL/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/immunology , Osteogenesis/genetics , Osteoporosis/etiology , Osteoporosis/genetics , Oxidation-Reduction , Single-Chain Antibodies/genetics
5.
JCI Insight ; 2(17)2017 09 07.
Article in English | MEDLINE | ID: mdl-28878136

ABSTRACT

Decreased cortical thickness and increased cortical porosity are the key anatomic changes responsible for osteoporotic fractures in elderly women and men. The cellular basis of these changes is unbalanced endosteal and intracortical osteonal remodeling by the osteoclasts and osteoblasts that comprise the basic multicellular units (BMUs). Like humans, mice lose cortical bone with age, but unlike humans, this loss occurs in the face of sex steroid sufficiency. Mice are therefore an ideal model to dissect age-specific osteoporotic mechanisms. Nevertheless, lack of evidence for endosteal or intracortical remodeling in mice has raised questions about their translational relevance. We show herein that administration of the antiosteoclastogenic cytokine osteoprotegerin to Swiss Webster mice ablated not only osteoclasts, but also endosteal bone formation, demonstrating the occurrence of BMU-based endosteal remodeling. Femoral cortical thickness decreased in aged male and female C57BL/6J mice, as well as F1 hybrids of C57BL/6J and BALB/cBy mice. This decrease was greater in C57BL/6J mice, indicating a genetic influence. Moreover, endosteal remodeling became unbalanced because of increased osteoclast and decreased osteoblast numbers. The porosity of the femoral cortex increased with age but was much higher in females of both strains. Notably, the increased cortical porosity resulted from de novo intracortical remodeling by osteon-like structures. Age-dependent cortical bone loss was associated with increased osteocyte DNA damage, cellular senescence, the senescence-associated secretory phenotype, and increased levels of RANKL. The demonstration of unbalanced endosteal and intracortical remodeling in old mice validates the relevance of this animal model to involutional osteoporosis in humans.


Subject(s)
Aging/pathology , Bone Remodeling , Porosity , Animals , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Osteoblasts/cytology , Osteoclasts/cytology
6.
Endocrinology ; 158(11): 3817-3831, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938402

ABSTRACT

In search of the sequence of pathogenic events leading to glucocorticoid-induced osteonecrosis, we determined the molecular, biomechanical, cellular, and vascular changes in the femur of C57BL/6 mice receiving prednisolone for 14, 28, or 42 days. The femoral head, but not the distal femur, of mice treated for 14 days showed a decrease in the expression of the hypoxia-inducible factor (Hif)-1α and vascular endothelial growth factor (VEGF), the number of osteoblasts, and bone formation rate and strength and showed an increase in osteoclasts. These changes were accompanied by conversion of the normal dendritic vasculature to pools of edema as detected by magnetic resonance imaging, providing robust diagnostic evidence of early osteonecrosis. At that time point, there were no detectable changes in bone density, cortical or cancellous bone architecture, midshaft or distal cancellous bone, or osteocyte apoptosis. In mice treated for 28 days, femoral head cancellous density, cortical width, and trabecular thickness decreased, and by 42 days the femoral heads had full-depth cortical penetrations and cancellous tissue osteonecrosis. These results indicate that the femoral head is a particularly sensitive anatomical site to the adverse effects of glucocorticoid excess on bone and that decreases of Hif-1α and VEGF expression, bone vascularity, and strength precede the loss of bone mass and microarchitectural deterioration, thus rendering the femoral head vulnerable to collapse.


Subject(s)
Femur Head Necrosis/chemically induced , Femur Head/drug effects , Glucocorticoids/adverse effects , Osteonecrosis/chemically induced , Animals , Apoptosis/drug effects , Bone Density/drug effects , Femur Head/pathology , Male , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/physiology , Osteogenesis/drug effects
7.
Am J Physiol Endocrinol Metab ; 310(9): E762-73, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26956187

ABSTRACT

ApoE-null (ApoE-KO) mice fed a high-fat diet (HFD) develop atherosclerosis, due in part to activation of vascular inflammation by oxidized low-density lipoprotein. Since bone loss also occurs in these mice, we used them to investigate the impact of oxidized lipids on bone homeostasis and to search for underlying pathogenic pathways. Four-month-old female ApoE-KO mice fed a HFD for three months exhibited increased levels of oxidized lipids in bone, as well as decreased femoral and vertebral trabecular and cortical bone mass, compared with ApoE-KO mice on normal diet. Despite HFD-induced increase in expression of Alox15, a lipoxygenase that oxidizes LDL and promotes atherogenesis, global deletion of this gene failed to ameliorate the skeletal impact of HFD. Osteoblast number and function were dramatically reduced in trabecular and cortical bone of HFD-fed mice, whereas osteoclast number was modestly reduced only in trabecular bone, indicating that an imbalance in favor of osteoclasts was responsible for HFD-induced bone loss. These changes were associated with decreased osteoblast progenitors and increased monocyte/macrophages in the bone marrow as well as increased expression of IL-1ß, IL-6, and TNF. HFD also attenuated Wnt signaling as evidenced by reduced expression of Wnt target genes, and it decreased expression of pro-osteoblastogenic Wnt ligands. These results suggest that oxidized lipids decrease bone mass by increasing anti-osteoblastogenic inflammatory cytokines and decreasing pro-osteoblastogenic Wnt ligands.


Subject(s)
Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Bone Diseases, Metabolic/genetics , Bone and Bones/immunology , Osteogenesis , Wnt Proteins/genetics , Absorptiometry, Photon , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Blotting, Western , Bone Density , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/immunology , Bone Diseases, Metabolic/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Bone and Bones/pathology , Cancellous Bone/diagnostic imaging , Cancellous Bone/immunology , Cancellous Bone/metabolism , Cancellous Bone/pathology , Cell Count , Cortical Bone/drug effects , Cortical Bone/immunology , Cortical Bone/metabolism , Cortical Bone/pathology , Diet, High-Fat , Female , Femur/diagnostic imaging , Femur/immunology , Femur/metabolism , Femur/pathology , Flow Cytometry , Immunomagnetic Separation , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lipoproteins, LDL/metabolism , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/immunology , Osteoblasts/cytology , Osteoclasts/cytology , Porosity , Reverse Transcriptase Polymerase Chain Reaction , Spine/diagnostic imaging , Spine/immunology , Spine/metabolism , Spine/pathology , Tumor Necrosis Factor-alpha/genetics
8.
J Bone Miner Res ; 31(4): 864-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26496249

ABSTRACT

Rac1 and Rac2 are thought to have important roles in osteoclasts. Therefore, mice with deletion of both Rac1 and Rac2 in mature osteoclasts (DKO) were generated by crossing Rac1(flox/flox) mice with mice expressing Cre in the cathepsin K locus and then mating these animals with Rac2(-/-) mice. DKO mice had markedly impaired tooth eruption. Bone mineral density (BMD) was increased 21% to 33% in 4- to 6-week-old DKO mice at all sites when measured by dual-energy X-ray absorptiometry (DXA) and serum cross-linked C-telopeptide (CTx) was reduced by 52%. The amount of metaphyseal trabecular bone was markedly increased in DKO mice, but the cortices were very thin. Spinal trabecular bone mass was increased. Histomorphometry revealed significant reductions in both osteoclast and osteoblast number and function in 4- to 6-week-old DKO animals. In 14- to 16-week-old animals, osteoclast number was increased, although bone density was further increased. DKO osteoclasts had severely impaired actin ring formation, an impaired ability to generate acid, and reduced resorptive activity in vitro. In addition, their life span ex vivo was reduced. DKO osteoblasts expressed normal differentiation markers except for the expression of osterix, which was reduced. The DKO osteoblasts mineralized normally in vitro, indicating that the in vivo defect in osteoblast function was not cell autonomous. Confocal imaging demonstrated focal disruption of the osteocytic dendritic network in DKO cortical bone. Despite these changes, DKO animals had a normal response to treatment with once-daily parathyroid hormone (PTH). We conclude that Rac1 and Rac2 have critical roles in skeletal metabolism.


Subject(s)
Aging , Gene Deletion , Neuropeptides , Osteoblasts , Osteoclasts , Osteopetrosis , rac GTP-Binding Proteins , rac1 GTP-Binding Protein , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Cell Count , Humans , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , RAC2 GTP-Binding Protein
9.
J Bone Miner Res ; 30(7): 1138-49, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25704845

ABSTRACT

In men, androgens are critical for the acquisition and maintenance of bone mass in both the cortical and cancellous bone compartment. Male mice with targeted deletion of the androgen receptor (AR) in mature osteoblasts or osteocytes have lower cancellous bone mass, but no cortical bone phenotype. We have investigated the possibility that the effects of androgens on the cortical compartment result from AR signaling in osteoprogenitors or cells of the osteoclast lineage; or via estrogen receptor alpha (ERα) signaling in either or both of these two cell types upon conversion of testosterone to estradiol. To this end, we generated mice with targeted deletion of an AR or an ERα allele in the mesenchymal (AR(f/y);Prx1-Cre or ERα(f/f);Osx1-Cre) or myeloid cell lineage (AR(f/y);LysM-Cre or ERα(f/f);LysM-Cre) and their descendants. Male AR(f/y);Prx1-Cre mice exhibited decreased bone volume and trabecular number, and increased osteoclast number in the cancellous compartment. Moreover, they did not undergo the loss of cancellous bone volume and trabecular number caused by orchidectomy (ORX) in their littermate controls. In contrast, AR(f/y);LysM-Cre, ERα(f/f);Osx1-Cre, or ERα(f/f);LysM-Cre mice had no cancellous bone phenotype at baseline and lost the same amount of cancellous bone as their controls following ORX. Most unexpectedly, adult males of all four models had no discernible cortical bone phenotype at baseline, and lost the same amount of cortical bone as their littermate controls after ORX. Recapitulation of the effects of ORX by AR deletion only in the AR(f/y);Prx1-Cre mice indicates that the effects of androgens on cancellous bone result from AR signaling in osteoblasts-not on osteoclasts or via aromatization. The effects of androgens on cortical bone mass, on the other hand, do not require AR or ERα signaling in any cell type across the osteoblast or osteoclast differentiation lineage. Therefore, androgens must exert their effects indirectly by actions on some other cell type(s) or tissue(s).


Subject(s)
Androgens/pharmacology , Bone and Bones/metabolism , Estrogen Receptor alpha/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Animals , Bone and Bones/drug effects , Cell Lineage/drug effects , Femur/diagnostic imaging , Femur/drug effects , Gene Deletion , Integrases/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , Orchiectomy , Organ Size/drug effects , Osteoblasts/drug effects , Osteoclasts/drug effects , Phenotype , X-Ray Microtomography
10.
Bone ; 75: 18-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25700544

ABSTRACT

Glucocorticoid excess decreases bone mass and strength in part by acting directly on osteoblasts and osteocytes, but the mechanisms remain unclear. Macroautophagy (herein referred to as autophagy) is a lysosome-based recycling pathway that promotes the turnover of intracellular components and can promote cell function and survival under stressful conditions. Recent studies have shown that glucocorticoids stimulate autophagy in osteocytes, suggesting that autophagy may oppose the negative actions of glucocorticoids on this cell type. To address this possibility, we compared the impact of prednisolone administration on the skeletons of adult mice in which autophagy was suppressed in osteocytes, via deletion of Atg7 with a Dmp1-Cre transgene, to their control littermates. In control mice, prednisolone increased autophagic flux in osteocyte-enriched bone as measured by LC3 conversion, but this change did not occur in the mice lacking Atg7 in osteocytes. Nonetheless, prednisolone reduced femoral cortical thickness, increased cortical porosity, and reduced bone strength to similar extents in mice with and without autophagy in osteocytes. Prednisolone also suppressed osteoblast number and bone formation in the cancellous bone of control mice. As shown previously, Atg7 deletion in osteocytes reduced osteoblast number and bone formation in cancellous bone, but these parameters were not further reduced by prednisolone administration. In cortical bone, prednisolone elevated osteoclast number to a similar extent in both genotypes. Taken together, these results demonstrate that although glucocorticoids stimulate autophagy in osteocytes, suppression of autophagy in this cell type does not worsen the negative impact of glucocorticoids on the skeleton.


Subject(s)
Autophagy/physiology , Bone and Bones/drug effects , Bone and Bones/metabolism , Glucocorticoids/adverse effects , Glucocorticoids/metabolism , Osteocytes/metabolism , Animals , Bone Density/drug effects , Bone and Bones/pathology , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteogenesis/drug effects , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
11.
Bone ; 66: 146-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24933342

ABSTRACT

Parathyroid hormone (PTH) excess stimulates bone resorption. This effect is associated with increased expression of the osteoclastogenic cytokine receptor activator of nuclear factor κB ligand (RANKL) in bone. However, several different cell types, including bone marrow stromal cells, osteocytes, and T lymphocytes, express both RANKL and the PTH receptor and it is unclear whether RANKL expression by any of these cell types is required for PTH-induced bone loss. Here we have used mice lacking the RANKL gene in osteocytes to determine whether RANKL produced by this cell type is required for the bone loss caused by secondary hyperparathyroidism induced by dietary calcium deficiency in adult mice. Thirty days of dietary calcium deficiency caused bone loss in control mice, but this effect was blunted in mice lacking RANKL in osteocytes. The increase in RANKL expression in bone and the increase in osteoclast number caused by dietary calcium deficiency were also blunted in mice lacking RANKL in osteocytes. These results demonstrate that RANKL produced by osteocytes contributes to the increased bone resorption and the bone loss caused by secondary hyperparathyroidism, strengthening the evidence that osteocytes are an important target cell for hormonal control of bone remodeling.


Subject(s)
Bone Resorption/metabolism , Bone Resorption/pathology , Calcium, Dietary/metabolism , Calcium/deficiency , Osteocytes/metabolism , Osteocytes/pathology , RANK Ligand/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Gene Deletion , Hyperthyroidism/complications , Hyperthyroidism/metabolism , Hyperthyroidism/pathology , Integrases/metabolism , Mice, Inbred C57BL
12.
Nat Commun ; 5: 3773, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24781012

ABSTRACT

Besides their cell-damaging effects in the setting of oxidative stress, reactive oxygen species (ROS) play an important role in physiological intracellular signalling by triggering proliferation and survival. FoxO transcription factors counteract ROS generation by upregulating antioxidant enzymes. Here we show that intracellular H2O2 accumulation is a critical and purposeful adaptation for the differentiation and survival of osteoclasts, the bone cells responsible for the resorption of mineralized bone matrix. Using mice with conditional loss or gain of FoxO transcription factor function, or mitochondria-targeted catalase in osteoclasts, we demonstrate this is achieved, at least in part, by downregulating the H2O2-inactivating enzyme catalase. Catalase downregulation results from the repression of the transcriptional activity of FoxO1, 3 and 4 by RANKL, the indispensable signal for the generation of osteoclasts, via an Akt-mediated mechanism. Notably, mitochondria-targeted catalase prevented the loss of bone caused by loss of oestrogens, suggesting that decreasing H2O2 production in mitochondria may represent a rational pharmacotherapeutic approach to diseases with increased bone resorption.


Subject(s)
Bone Resorption/physiopathology , Catalase/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic/physiology , Hydrogen Peroxide/metabolism , Reactive Oxygen Species/metabolism , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , DNA Primers/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Enzymologic/genetics , Mice , RANK Ligand/metabolism , Real-Time Polymerase Chain Reaction , X-Ray Microtomography
13.
J Bone Miner Res ; 29(1): 1-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23712442

ABSTRACT

Bisphosphonates (BPs) and denosumab reduce the risk of spine and nonspine fractures. Atypical femur fractures (AFFs) located in the subtrochanteric region and diaphysis of the femur have been reported in patients taking BPs and in patients on denosumab, but they also occur in patients with no exposure to these drugs. In this report, we review studies on the epidemiology, pathogenesis, and medical management of AFFs, published since 2010. This newer evidence suggests that AFFs are stress or insufficiency fractures. The original case definition was revised to highlight radiographic features that distinguish AFFs from ordinary osteoporotic femoral diaphyseal fractures and to provide guidance on the importance of their transverse orientation. The requirement that fractures be noncomminuted was relaxed to include minimal comminution. The periosteal stress reaction at the fracture site was changed from a minor to a major feature. The association with specific diseases and drug exposures was removed from the minor features, because it was considered that these associations should be sought rather than be included in the case definition. Studies with radiographic review consistently report significant associations between AFFs and BP use, although the strength of associations and magnitude of effect vary. Although the relative risk of patients with AFFs taking BPs is high, the absolute risk of AFFs in patients on BPs is low, ranging from 3.2 to 50 cases per 100,000 person-years. However, long-term use may be associated with higher risk (∼100 per 100,000 person-years). BPs localize in areas that are developing stress fractures; suppression of targeted intracortical remodeling at the site of an AFF could impair the processes by which stress fractures normally heal. When BPs are stopped, risk of an AFF may decline. Lower limb geometry and Asian ethnicity may contribute to the risk of AFFs. There is inconsistent evidence that teriparatide may advance healing of AFFs.


Subject(s)
Bone Density Conservation Agents/adverse effects , Diphosphonates/adverse effects , Femoral Fractures/diagnostic imaging , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Denosumab , Diaphyses/diagnostic imaging , Diaphyses/pathology , Female , Femoral Fractures/epidemiology , Fractures, Stress/diagnostic imaging , Fractures, Stress/epidemiology , Humans , Male , Middle Aged , Radiography , Risk Factors
14.
J Bone Miner Res ; 29(1): 103-17, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23761243

ABSTRACT

Skeletal aging is accompanied by decreased cancellous bone mass and increased formation of pores within cortical bone. The latter accounts for a large portion of the increase in nonvertebral fractures after age 65 years in humans. We selectively deleted Bak and Bax, two genes essential for apoptosis, in two types of terminally differentiated bone cells: the short-lived osteoblasts that elaborate the bone matrix, and the long-lived osteocytes that are immured within the mineralized matrix and choreograph the regeneration of bone. Attenuation of apoptosis in osteoblasts increased their working lifespan and thereby cancellous bone mass in the femur. In long-lived osteocytes, however, it caused dysfunction with advancing age and greatly magnified intracortical femoral porosity associated with increased production of receptor activator of nuclear factor-κB ligand and vascular endothelial growth factor. Increasing bone mass by artificial prolongation of the inherent lifespan of short-lived osteoblasts, while exaggerating the adverse effects of aging on long-lived osteocytes, highlights the seminal role of cell age in bone homeostasis. In addition, our findings suggest that distress signals produced by old and/or dysfunctional osteocytes are the culprits of the increased intracortical porosity in old age.


Subject(s)
Aging/pathology , Apoptosis/drug effects , Osteoblasts/physiology , Osteocytes/physiology , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency , Animals , Bone Density/drug effects , Bone Remodeling/physiology , Calcium/metabolism , Female , Femur/pathology , Femur/physiopathology , Homeostasis , Mice , Osteocytes/pathology , Porosity
15.
J Clin Invest ; 123(8): 3409-19, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867625

ABSTRACT

Wnt/ß-catenin/TCF signaling stimulates bone formation and suppresses adipogenesis. The hallmarks of skeletal involution with age, on the other hand, are decreased bone formation and increased bone marrow adiposity. These changes are associated with increased oxidative stress and decreased growth factor production, which activate members of the FOXO family of transcription factors. FOXOs in turn attenuate Wnt/ß-catenin signaling by diverting ß-catenin from TCF- to FOXO-mediated transcription. We show herein that mice lacking Foxo1, -3, and -4 in bipotential progenitors of osteoblast and adipocytes (expressing Osterix1) exhibited increased osteoblast number and high bone mass that was maintained in old age as well as decreased adiposity in the aged bone marrow. The increased bone mass in the Foxo-deficient mice was accounted for by increased proliferation of osteoprogenitor cells and bone formation resulting from upregulation of Wnt/ß-catenin signaling and cyclin D1 expression, but not changes in redox balance. Consistent with this mechanism, ß-catenin deletion in Foxo null cells abrogated both the increased cyclin D1 expression and proliferation. The elucidation of a restraining effect of FOXOs on Wnt signaling in bipotential progenitors suggests that FOXO activation by accumulation of age-associated cellular stressors may be a seminal pathogenetic mechanism in the development of involutional osteoporosis.


Subject(s)
Forkhead Transcription Factors/genetics , Osteogenesis , Wnt Signaling Pathway , Adipogenesis , Adiposity , Animals , Bone Density , Bone Marrow/anatomy & histology , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Female , Femur/cytology , Femur/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/deficiency , Gene Knockout Techniques , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Reactive Oxygen Species/metabolism , Sp7 Transcription Factor , Stem Cells/physiology , Transcription Factors/metabolism
16.
J Biol Chem ; 288(24): 17432-40, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23645674

ABSTRACT

Bone mass declines with age but the mechanisms responsible remain unclear. Here we demonstrate that deletion of a conditional allele for Atg7, a gene essential for autophagy, from osteocytes caused low bone mass in 6-month-old male and female mice. Cancellous bone volume and cortical thickness were decreased, and cortical porosity increased, in conditional knock-out mice compared with control littermates. These changes were associated with low osteoclast number, osteoblast number, bone formation rate, and wall width in the cancellous bone of conditional knock-out mice. In addition, oxidative stress was higher in the bones of conditional knock-out mice as measured by reactive oxygen species levels in the bone marrow and by p66(shc) phosphorylation in L6 vertebra. Each of these changes has been previously demonstrated in the bones of old versus young adult mice. Thus, these results demonstrate that suppression of autophagy in osteocytes mimics, in many aspects, the impact of aging on the skeleton and suggest that a decline in autophagy with age may contribute to the low bone mass associated with aging.


Subject(s)
Femur/metabolism , Lumbar Vertebrae/metabolism , Osteocytes/physiology , Aging , Animals , Autophagy , Autophagy-Related Protein 7 , Bone Density , Cell Differentiation , Cells, Cultured , Female , Femur/diagnostic imaging , Femur/pathology , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Osteoblasts/physiology , Osteoclasts/physiology , Oxidative Stress , Radiography , Reactive Oxygen Species/metabolism
17.
Mol Endocrinol ; 27(4): 649-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23443267

ABSTRACT

Extensive evidence has suggested that at least some of the effects of estrogens on bone are mediated via extranuclear estrogen receptor α signaling. However, definitive proof for this contention and the extent to which such effects may contribute to the overall protective effects of estrogens on bone maintenance have remained elusive. Here, we investigated the ability of a 17ß-estradiol (E2) dendrimer conjugate (EDC), incapable of stimulating nuclear-initiated actions of estrogen receptor α, to prevent the effects of ovariectomy (OVX) on the murine skeleton. We report that EDC was as potent as an equimolar dose of E2 in preventing bone loss in the cortical compartment that represents 80% of the entire skeleton, but was ineffective on cancellous bone. In contrast, E2 was effective in both compartments. Consistent with its effect on cortical bone mass, EDC partially prevented the loss of both vertebral and femoral strength. In addition, EDC, as did E2, prevented the OVX-induced increase in osteoclastogenesis, osteoblastogenesis, and oxidative stress. Nonetheless, the OVX-induced decrease in uterine weight was unaltered by EDC but was restored by E2. These results demonstrate that the protection of cortical bone mass by estrogens is mediated, at least in part, via a mechanism that is distinct from the classic mechanism of estrogen action on reproductive organs.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/pathology , Cell Nucleus/metabolism , Estrogen Receptor alpha/metabolism , Animals , Atrophy , Bone Density/drug effects , Bone Remodeling/drug effects , Bone and Bones/drug effects , Cell Nucleus/drug effects , Estradiol/pharmacology , Female , Femur/drug effects , Femur/pathology , Femur/physiopathology , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , Ovariectomy , Oxidative Stress/drug effects , Spine/drug effects , Spine/pathology , Spine/physiopathology , Uterus/drug effects , Uterus/pathology
18.
Bone ; 54(2): 264-71, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23238124

ABSTRACT

Apoptotic death of osteocytes was recognized over 15 years ago, but its significance for bone homeostasis has remained elusive. A new paradigm has emerged that invokes osteocyte apoptosis as a critical event in the recruitment of osteoclasts to a specific site in response to skeletal unloading, fatigue damage, estrogen deficiency and perhaps in other states where bone must be removed. This is accomplished by yet to be defined signals emanating from dying osteocytes, which stimulate neighboring viable osteocytes to produce osteoclastogenic cytokines. The osteocyte apoptosis caused by chronic glucocorticoid administration does not increase osteoclasts; however, it does negatively impact maintenance of bone hydration, vascularity, and strength.


Subject(s)
Apoptosis , Osteocytes/cytology , Animals , Bone Remodeling , Humans , Osteocytes/metabolism , Osteocytes/ultrastructure , RANK Ligand/metabolism , Signal Transduction , Weight-Bearing
19.
J Clin Invest ; 123(1): 394-404, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23221342

ABSTRACT

The detection of estrogen receptor-α (ERα) in osteoblasts and osteoclasts over 20 years ago suggested that direct effects of estrogens on both of these cell types are responsible for their beneficial effects on the skeleton, but the role of ERα in osteoblast lineage cells has remained elusive. In addition, estrogen activation of ERα in osteoclasts can only account for the protective effect of estrogens on the cancellous, but not the cortical, bone compartment that represents 80% of the entire skeleton. Here, we deleted ERα at different stages of differentiation in murine osteoblast lineage cells. We found that ERα in osteoblast progenitors expressing Osterix1 (Osx1) potentiates Wnt/ß-catenin signaling, thereby increasing proliferation and differentiation of periosteal cells. Further, this signaling pathway was required for optimal cortical bone accrual at the periosteum in mice. Notably, this function did not require estrogens. The osteoblast progenitor ERα mediated a protective effect of estrogens against endocortical, but not cancellous, bone resorption. ERα in mature osteoblasts or osteocytes did not influence cancellous or cortical bone mass. Hence, the ERα in both osteoblast progenitors and osteoclasts functions to optimize bone mass but at distinct bone compartments and in response to different cues.


Subject(s)
Cell Differentiation/physiology , Estrogen Receptor alpha/metabolism , Osteoblasts/metabolism , Periosteum/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Cell Proliferation , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Mice , Mice, Mutant Strains , Osteoblasts/cytology , Osteoclasts/cytology , Osteoclasts/metabolism , Periosteum/cytology , Sp7 Transcription Factor , Stem Cells/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics
20.
Endocrinol Metab Clin North Am ; 41(3): 595-611, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22877431

ABSTRACT

Glucocorticoid administration is the most common cause of secondary osteoporosis and the leading cause of nontraumatic osteonecrosis. In patients receiving long-term therapy, glucocorticoids induce fractures in 30% to 50% and osteonecrosis in 9% to 40%. This article reviews glucocorticoid-induced osteoporosis and osteonecrosis, addressing the risk factors, pathogenesis, evaluation, treatment, and uncertainties in the clinical management of these disorders.


Subject(s)
Bone and Bones/drug effects , Glucocorticoids/adverse effects , Osteonecrosis/chemically induced , Osteoporosis/chemically induced , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Apoptosis/drug effects , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Bone and Bones/metabolism , Denosumab , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Drug Monitoring , Female , Humans , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocytes/drug effects , Osteocytes/metabolism , Osteonecrosis/drug therapy , Osteonecrosis/physiopathology , Osteoporosis/drug therapy , Osteoporosis/physiopathology , Osteoporotic Fractures/etiology , Osteoporotic Fractures/prevention & control , Teriparatide/pharmacology , Teriparatide/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...