Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 37(9): 1750-1760, 2022 09.
Article in English | MEDLINE | ID: mdl-35789113

ABSTRACT

Estrogen is known to regulate bone metabolism in both women and men, but substantial gaps remain in our knowledge of estrogen and estrogen receptor alpha (ERα) regulation of adult bone metabolism. Studies using global ERα-knockout mice were confounded by high circulating sex-steroid levels, and osteocyte/osteoblast-specific ERα deletion may be confounded by ERα effects on growth versus the adult skeleton. Thus, we developed mice expressing the tamoxifen-inducible CreERT2 in osteocytes using the 8-kilobase (kb) Dmp1 promoter (Dmp1CreERT2 ). These mice were crossed with ERαfl//fl mice to create ERαΔOcy mice, permitting inducible osteocyte-specific ERα deletion in adulthood. After intermittent tamoxifen treatment of adult 4-month-old mice for 1 month, female, but not male, ERαΔOcy mice exhibited reduced spine bone volume fraction (BV/TV (-20.1%, p = 0.004) accompanied by decreased trabecular bone formation rate (-18.9%, p = 0.0496) and serum P1NP levels (-38.9%, p = 0.014). Periosteal (+65.6%, p = 0.004) and endocortical (+64.1%, p = 0.003) expansion were higher in ERαΔOcy mice compared to control (Dmp1CreERT2 ) mice at the tibial diaphysis, reflecting the known effects of estrogen to inhibit periosteal apposition and promote endocortical formation. Increases in Sost (2.1-fold, p = 0.001) messenger RNA (mRNA) levels were observed in trabecular bone at the spine in ERαΔOcy mice, consistent with previous reports that estrogen deficiency is associated with increased circulating sclerostin as well as bone SOST mRNA levels in humans. Further, the biological consequences of increased Sost expression were reflected in significant overall downregulation in panels of osteoblast and Wnt target genes in osteocyte-enriched bones from ERαΔOcy mice. These findings thus establish that osteocytic ERα is critical for estrogen action in female, but not male, adult bone metabolism. Moreover, the reduction in bone formation accompanied by increased Sost, decreased osteoblast, and decreased Wnt target gene expression in ERαΔOcy mice provides a direct link in vivo between ERα and Wnt signaling. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Estrogen Receptor alpha , Osteocytes , Adaptor Proteins, Signal Transducing/metabolism , Adult , Animals , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Female , Humans , Infant , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteocytes/metabolism , RNA, Messenger/metabolism , Tamoxifen/pharmacology
2.
Proc Natl Acad Sci U S A ; 119(26): e2201490119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35733270

ABSTRACT

Excess bone loss due to increased osteoclastogenesis is a significant clinical problem. Intraflagellar transport (IFT) proteins have been reported to regulate cell growth and differentiation. The role of IFT80, an IFT complex B protein, in osteoclasts (OCs) is completely unknown. Here, we demonstrate that deletion of IFT80 in the myeloid lineage led to increased OC formation and activity accompanied by severe bone loss in mice. IFT80 regulated OC formation by associating with Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b) to promote protein stabilization and proteasomal degradation of tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6). IFT80 knockdown resulted in increased ubiquitination of Cbl-b and higher TRAF6 levels, thereby hyperactivating the receptor activator of nuclear factor-κß (NF-κß) ligand (RANKL) signaling axis and increased OC formation. Ectopic overexpression of IFT80 rescued osteolysis in a calvarial model of bone loss. We have thus identified a negative function of IFT80 in OCs.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Resorption , Carrier Proteins , Osteoclasts , Osteogenesis , Proto-Oncogene Proteins c-cbl , TNF Receptor-Associated Factor 6 , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Resorption/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Disease Models, Animal , Gene Deletion , Mice , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis/genetics , Proteolysis , Proto-Oncogene Proteins c-cbl/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination
3.
iScience ; 23(6): 101172, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32512384

ABSTRACT

Rheumatoid arthritis (RA) is the most common inflammatory disease, which currently lacks effective treatment. Here, we discovered that the Regulator of G Protein Signaling 12 (RGS12) plays a key role in regulating inflammation. Transcriptional and protein analysis revealed that RGS12 was upregulated in human and mouse RA macrophages. Deletion of RGS12 in myeloid lineage or globally inhibits the development of collagen-induced arthritis including joint swelling and bone destruction. Mechanistically, RGS12 associates with NF-κB(p65) to activate its phosphorylation and nuclear translocation through PTB domain, and NF-κB(p65) regulates RGS12 expression in a transcriptional manner. The nuclear translocation ability of NF-κB(p65) and RGS12 can both be enhanced by cyclooxygenase-2 (COX2). Furthermore, ablation of RGS12 via RNA interference significantly blocks the inflammatory process in vivo and in vitro. These results demonstrate that RGS12 plays a critical role in the pathogenesis of inflammatory arthritis.

4.
Nat Commun ; 11(1): 87, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31911667

ABSTRACT

Bone remodeling consists of resorption by osteoclasts followed by formation by osteoblasts, and osteoclasts are a source of bone formation-stimulating factors. Here we utilize osteoclast ablation by denosumab (DMAb) and RNA-sequencing of bone biopsies from postmenopausal women to identify osteoclast-secreted factors suppressed by DMAb. Based on these analyses, LIF, CREG2, CST3, CCBE1, and DPP4 are likely osteoclast-derived coupling factors in humans. Given the role of Dipeptidyl Peptidase-4 (DPP4) in glucose homeostasis, we further demonstrate that DMAb-treated participants have a significant reduction in circulating DPP4 and increase in Glucagon-like peptide (GLP)-1 levels as compared to the placebo-treated group, and also that type 2 diabetic patients treated with DMAb show significant reductions in HbA1c as compared to patients treated either with bisphosphonates or calcium and vitamin D. Thus, our results identify several coupling factors in humans and uncover osteoclast-derived DPP4 as a potential link between bone remodeling and energy metabolism.


Subject(s)
Bone and Bones/metabolism , Energy Metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Aged , Aged, 80 and over , Animals , Bone Remodeling , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Denosumab/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Energy Metabolism/drug effects , Female , Humans , Middle Aged , Osteoblasts/drug effects , Osteoclasts/drug effects , Prospective Studies , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
Elife ; 82019 09 06.
Article in English | MEDLINE | ID: mdl-31490121

ABSTRACT

Regulators of G-protein Signaling are a conserved family of proteins required in various biological processes including cell differentiation. We previously demonstrated that Rgs12 is essential for osteoclast differentiation and its deletion in vivo protected mice against pathological bone loss. To characterize its mechanism in osteoclastogenesis, we selectively deleted Rgs12 in C57BL/6J mice targeting osteoclast precursors using LyzM-driven Cre mice or overexpressed Rgs12 in RAW264.7 cells. Rgs12 deletion in vivo led to an osteopetrotic phenotype evidenced by increased trabecular bone, decreased osteoclast number and activity but no change in osteoblast number and bone formation. Rgs12 overexpression increased osteoclast number and size, and bone resorption activity. Proteomics analysis of Rgs12-depleted osteoclasts identified an upregulation of antioxidant enzymes under the transcriptional regulation of Nrf2, the master regulator of oxidative stress. We confirmed an increase of Nrf2 activity and impaired reactive oxygen species production in Rgs12-deficient cells. Conversely, Rgs12 overexpression suppressed Nrf2 through a mechanism dependent on the 26S proteasome, and promoted RANKL-induced phosphorylation of ERK1/2 and NFκB, which was abrogated by antioxidant treatment. Our study therefore identified a novel role of Rgs12 in regulating Nrf2, thereby controlling cellular redox state and osteoclast differentiation.


Subject(s)
Antioxidants/metabolism , NF-E2-Related Factor 2/metabolism , Osteogenesis , RGS Proteins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , RGS Proteins/deficiency
7.
J Biol Chem ; 294(31): 11772-11784, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31189651

ABSTRACT

Enhanced osteoclast-mediated bone resorption and diminished formation may promote bone loss. Pleckstrin homology (PH) domain and leucine-rich repeat protein phosphatase 1 (Phlpp1) regulates protein kinase C (PKC) and other proteins in the control of bone mass. Germline Phlpp1 deficiency reduces bone volume, but the mechanisms remain unknown. Here, we found that conditional Phlpp1 deletion in murine osteoclasts increases their numbers, but also enhances bone mass. Despite elevating osteoclasts, Phlpp1 deficiency did not increase serum markers of bone resorption, but elevated serum markers of bone formation. These results suggest that Phlpp1 suppresses osteoclast formation and production of paracrine factors controlling osteoblast activity. Phlpp1 deficiency elevated osteoclast numbers and size in ex vivo osteoclastogenesis assays, accompanied by enhanced expression of proto-oncogene C-Fms (C-Fms) and hyper-responsiveness to macrophage colony-stimulating factor (M-CSF) in bone marrow macrophages. Although Phlpp1 deficiency increased TRAP+ cell numbers, it suppressed actin-ring formation and bone resorption in these assays. We observed that Phlpp1 deficiency increases activity of PKCζ, a PKC isoform controlling cell polarity, and that addition of a PKCζ pseudosubstrate restores osteoclastogenesis and bone resorption of Phlpp1-deficient osteoclasts. Moreover, Phlpp1 deficiency increased expression of the bone-coupling factor collagen triple helix repeat-containing 1 (Cthrc1). Conditioned growth medium derived from Phlpp1-deficient osteoclasts enhanced mineralization of ex vivo osteoblast cultures, an effect that was abrogated by Cthrc1 knockdown. In summary, Phlpp1 critically regulates osteoclast numbers, and Phlpp1 deficiency enhances bone mass despite higher osteoclast numbers because it apparently disrupts PKCζ activity, cell polarity, and bone resorption and increases secretion of bone-forming Cthrc1.


Subject(s)
Osteogenesis , Phosphoprotein Phosphatases/metabolism , Animals , Bone Density , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/physiology , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/metabolism , Female , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice , Mice, Knockout , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis/drug effects , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Protein Kinase C/metabolism , RNA Interference , RNA, Small Interfering/metabolism
9.
Nat Med ; 23(9): 1072-1079, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28825716

ABSTRACT

Aging is associated with increased cellular senescence, which is hypothesized to drive the eventual development of multiple comorbidities. Here we investigate a role for senescent cells in age-related bone loss through multiple approaches. In particular, we used either genetic (i.e., the INK-ATTAC 'suicide' transgene encoding an inducible caspase 8 expressed specifically in senescent cells) or pharmacological (i.e., 'senolytic' compounds) means to eliminate senescent cells. We also inhibited the production of the proinflammatory secretome of senescent cells using a JAK inhibitor (JAKi). In aged (20- to 22-month-old) mice with established bone loss, activation of the INK-ATTAC caspase 8 in senescent cells or treatment with senolytics or the JAKi for 2-4 months resulted in higher bone mass and strength and better bone microarchitecture than in vehicle-treated mice. The beneficial effects of targeting senescent cells were due to lower bone resorption with either maintained (trabecular) or higher (cortical) bone formation as compared to vehicle-treated mice. In vitro studies demonstrated that senescent-cell conditioned medium impaired osteoblast mineralization and enhanced osteoclast-progenitor survival, leading to increased osteoclastogenesis. Collectively, these data establish a causal role for senescent cells in bone loss with aging, and demonstrate that targeting these cells has both anti-resorptive and anabolic effects on bone. Given that eliminating senescent cells and/or inhibiting their proinflammatory secretome also improves cardiovascular function, enhances insulin sensitivity, and reduces frailty, targeting this fundamental mechanism to prevent age-related bone loss suggests a novel treatment strategy not only for osteoporosis, but also for multiple age-related comorbidities.


Subject(s)
Bone and Bones/drug effects , Cellular Senescence/drug effects , Janus Kinases/antagonists & inhibitors , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteocytes/drug effects , Osteoporosis/metabolism , Pyrazoles/pharmacology , Absorptiometry, Photon , Animals , Apoptosis/genetics , Bone and Bones/metabolism , Cancellous Bone/drug effects , Cancellous Bone/metabolism , Caspase 8/genetics , Cell Differentiation , Cellular Senescence/genetics , Cortical Bone/drug effects , Cortical Bone/metabolism , Culture Media, Conditioned , Flow Cytometry , Gene Expression Profiling , In Vitro Techniques , Mice , Mice, Transgenic , Nitriles , Osteoblasts/cytology , Osteoclasts/cytology , Osteoporosis/genetics , Pyrimidines , Real-Time Polymerase Chain Reaction , Weight-Bearing , beta-Galactosidase
10.
Endocr Rev ; 38(4): 325-350, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28651365

ABSTRACT

Cathepsin K is a cysteine protease member of the cathepsin lysosomal protease family. Although cathepsin K is highly expressed in osteoclasts, lower levels of cathepsin K are also found in a variety of other tissues. Secretion of cathepsin K from the osteoclast into the sealed osteoclast-bone cell interface results in efficient degradation of type I collagen. The absence of cathepsin K activity in humans results in pycnodysostosis, characterized by increased bone mineral density and fractures. Pharmacologic cathepsin K inhibition leads to continuous increases in bone mineral density for ≤5 years of treatment and improves bone strength at the spine and hip. Compared with other antiresorptive agents, cathepsin K inhibition is nearly equally efficacious for reducing biochemical markers of bone resorption but comparatively less active for reducing bone formation markers. Despite multiple efforts to develop cathepsin K inhibitors, potential concerns related to off-target effects of the inhibitors against other cathepsins and cathepsin K inhibition at nonbone sites, including skin and perhaps cardiovascular and cerebrovascular sites, prolonged the regulatory approval process. A large multinational randomized, double-blind phase III study of odanacatib in postmenopausal women with osteoporosis was recently completed. Although that study demonstrated clinically relevant reductions in fractures at multiple sites, odanacatib was ultimately withdrawn from the regulatory approval process after it was found to be associated with an increased risk of cerebrovascular accidents. Nonetheless, the underlying biology and clinical effects of cathepsin K inhibition remain of considerable interest and could guide future therapeutic approaches for osteoporosis.


Subject(s)
Cathepsin K/antagonists & inhibitors , Cathepsin K/metabolism , Enzyme Inhibitors/pharmacology , Osteoporosis/metabolism , Humans , Osteoporosis/drug therapy
11.
Bone ; 96: 45-50, 2017 03.
Article in English | MEDLINE | ID: mdl-27888056

ABSTRACT

Sclerostin, the product of the SOST gene, is a secreted inhibitor of Wnt signaling that is produced by osteocytes to regulate bone formation. While it is often considered an osteocyte-specific protein, SOST expression has been reported in numerous other cell types, including hypertrophic chondrocytes and cementocytes. Of interest, SOST/sclerostin expression is altered in certain pathogenic conditions, including osteoarthritis and rheumatic joint disease, and it is unclear whether sclerostin plays a protective role or whether sclerostin may mediate disease pathogenesis. Therefore, as anti-sclerostin antibodies are being developed for the treatment of osteoporosis, it is important to understand the functions of sclerostin beyond the regulation of bone formation.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Osteocytes/metabolism , Animals , Disease , Humans , Organ Specificity
12.
Sci Signal ; 9(440): ra79, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27507649

ABSTRACT

Histone deacetylase (HDAC) inhibitors are efficacious epigenetic-based therapies for some cancers and neurological disorders; however, each of these drugs inhibits multiple HDACs and has detrimental effects on the skeleton. To better understand how HDAC inhibitors affect endochondral bone formation, we conditionally deleted one of their targets, Hdac3, pre- and postnatally in type II collagen α1 (Col2α1)-expressing chondrocytes. Embryonic deletion was lethal, but postnatal deletion of Hdac3 delayed secondary ossification center formation, altered maturation of growth plate chondrocytes, and increased osteoclast activity in the primary spongiosa. HDAC3-deficient chondrocytes exhibited increased expression of cytokine and matrix-degrading genes (Il-6, Mmp3, Mmp13, and Saa3) and a reduced abundance of genes related to extracellular matrix production, bone development, and ossification (Acan, Col2a1, Ihh, and Col10a1). Histone acetylation increased at and near genes that had increased expression. The acetylation and activation of nuclear factor κB (NF-κB) were also increased in HDAC3-deficient chondrocytes. Increased cytokine signaling promoted autocrine activation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and NF-κB pathways to suppress chondrocyte maturation, as well as paracrine activation of osteoclasts and bone resorption. Blockade of interleukin-6 (IL-6)-JAK-STAT signaling, NF-κB signaling, and bromodomain extraterminal proteins, which recognize acetylated lysines and promote transcriptional elongation, significantly reduced Il-6 and Mmp13 expression in HDAC3-deficient chondrocytes and secondary activation in osteoclasts. The JAK inhibitor ruxolitinib also reduced osteoclast activity in Hdac3 conditional knockout mice. Thus, HDAC3 controls the temporal and spatial expression of tissue-remodeling genes and inflammatory responses in chondrocytes to ensure proper endochondral ossification during development.


Subject(s)
Autocrine Communication/physiology , Extracellular Matrix/metabolism , Histone Deacetylases/metabolism , Interleukin-6/metabolism , Osteogenesis/physiology , Signal Transduction/physiology , Animals , Autocrine Communication/drug effects , Chondrocytes/metabolism , Extracellular Matrix/genetics , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Histone Deacetylases/genetics , Interleukin-6/genetics , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Mice, Knockout , Nitriles , Osteoclasts/metabolism , Osteogenesis/drug effects , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction/drug effects
13.
J Bone Miner Res ; 31(1): 116-28, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26211746

ABSTRACT

Bone loss and increased marrow adiposity are hallmarks of aging skeletons. Conditional deletion of histone deacetylase 3 (Hdac3) in murine osteochondroprogenitor cells causes osteopenia and increases marrow adiposity, even in young animals, but the origins of the increased adiposity are unclear. To explore this, bone marrow stromal cells (BMSCs) from Hdac3-depleted and control mice were cultured in osteogenic medium. Hdac3-deficient cultures accumulated lipid droplets in greater abundance than control cultures and expressed high levels of genes related to lipid storage (Fsp27/Cidec, Plin1) and glucocorticoid metabolism (Hsd11b1) despite normal levels of Pparγ2. Approximately 5% of the lipid containing cells in the wild-type cultures expressed the master osteoblast transcription factor Runx2, but this population was threefold greater in the Hdac3-depleted cultures. Adenoviral expression of Hdac3 restored normal gene expression, indicating that Hdac3 controls glucocorticoid activation and lipid storage within osteoblast lineage cells. HDAC3 expression was reduced in bone cells from postmenopausal as compared to young women, and in osteoblasts from aged as compared to younger mice. Moreover, phosphorylation of S424 in Hdac3, a posttranslational mark necessary for deacetylase activity, was suppressed in osseous cells from old mice. Thus, concurrent declines in transcription and phosphorylation combine to suppress Hdac3 activity in aging bone, and reduced Hdac3 activity in osteochondroprogenitor cells contributes to increased marrow adiposity associated with aging. © 2015 American Society for Bone and Mineral Research.


Subject(s)
Adiposity , Aging , Bone Marrow Cells/metabolism , Glucocorticoids/metabolism , Histone Deacetylases/deficiency , Lipid Metabolism , Stem Cells/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Female , Glucocorticoids/genetics , Histone Deacetylases/metabolism , Humans , Mice , Mice, Transgenic , PPAR gamma/genetics , PPAR gamma/metabolism , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteins/genetics , Proteins/metabolism , Stem Cells/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
14.
J Bone Miner Res ; 31(1): 65-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26189772

ABSTRACT

Although there has been extensive characterization of the Wnt signaling pathway in the osteoblast lineage, the effects of Wnt proteins on the osteoclast lineage are less well studied. We found that osteoclast lineage cells express canonical Wnt receptors. Wnt3a reduced osteoclast formation when applied to early bone-marrow macrophage (BMM) osteoclast differentiation cultures, whereas late addition did not suppress osteoclast formation. Early Wnt3a treatment inactivated the crucial transcription factor NFATc1 in osteoclast progenitors. Wnt3a led to the accumulation of nuclear ß-catenin, confirming activation of canonical Wnt signaling. Reducing low-density lipoprotein receptor-related proteins (Lrp) 5 and Lrp6 protein expression prevented Wnt3a-induced inactivation of NFATc1; however, deletion of ß-catenin did not block Wnt3a inactivation of NFATc1, suggesting that this effect was mediated by a noncanonical pathway. Wnt3a rapidly activated the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway and pharmacological stimulation of cAMP/PKA signaling suppressed osteoclast differentiation; Wnt3a-induced NFATc1 phosphorylation was blocked by inhibiting interactions between PKA and A-kinase anchoring proteins (AKAPs). These data indicate that Wnt3a directly suppresses osteoclast differentiation through both canonical (ß-catenin) and noncanonical (cAMP/PKA) pathways in osteoclast precursors. In vivo reduction of Lrp5 and Lrp6 expressions in the early osteoclast lineage via Rank promoter Cre recombination reduced trabecular bone mass, whereas disruption of Lrp5/6 expression in late osteoclast precursors via cathepsin K (Ctsk) promoter Cre recombination did not alter the skeletal phenotype. Surprisingly, reduction of Lrp5/6 in the early osteoclast lineage decreased osteoclast numbers, as well as osteoblast numbers. Published studies have previously noted that ß-catenin signaling is required for osteoclast progenitor proliferation. Our in vivo data suggest that Rank promoter Cre-mediated deletion of Lrp5/6 may similarly impair osteoclast progenitor proliferation.


Subject(s)
Cell Differentiation/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Osteoclasts/metabolism , Wnt3A Protein/metabolism , A Kinase Anchor Proteins/metabolism , Animals , Enzyme Activation/physiology , Low Density Lipoprotein Receptor-Related Protein-5/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-6/biosynthesis , Mice , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , beta Catenin/metabolism
15.
J Bone Miner Res ; 31(1): 76-85, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26108893

ABSTRACT

Osteoblast-mediated bone formation is coupled to osteoclast-mediated bone resorption. These processes become uncoupled with age, leading to increased risk for debilitating fractures. Therefore, understanding how osteoblasts are recruited to sites of resorption is vital to treating age-related bone loss. Osteoclasts release and activate TGF-ß from the bone matrix. Here we show that osteoclast-specific inhibition of TGF-ß receptor signaling in mice results in osteopenia due to reduced osteoblast numbers with no significant impact on osteoclast numbers or activity. TGF-ß induced osteoclast expression of Wnt1, a protein crucial to normal bone formation, and this response was blocked by impaired TGF-ß receptor signaling. Osteoclasts in aged murine bones had lower TGF-ß signaling and Wnt1 expression in vivo. Ex vivo stimulation of osteoclasts derived from young or old mouse bone marrow macrophages showed no difference in TGF-ß-induced Wnt1 expression. However, young osteoclasts expressed reduced Wnt1 when cultured on aged mouse bone chips compared to young mouse bone chips, consistent with decreased skeletal TGF-ß availability with age. Therefore, osteoclast responses to TGF-ß are essential for coupling bone resorption to bone formation, and modulating this pathway may provide opportunities to treat age-related bone loss.


Subject(s)
Bone Resorption/metabolism , Osteoclasts/metabolism , Osteogenesis/physiology , Receptors, Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/physiology , Wnt1 Protein/metabolism , Animals , Bone Resorption/genetics , Gene Expression Regulation , Mice , Mice, Transgenic , Osteoclasts/physiology , Receptors, Transforming Growth Factor beta/genetics , Wnt1 Protein/genetics
17.
Curr Osteoporos Rep ; 12(1): 107-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24477413

ABSTRACT

The SOST gene, which encodes the protein sclerostin, was identified through genetic linkage analysis of sclerosteosis and van Buchem's disease patients. Sclerostin is a secreted glycoprotein that binds to the low-density lipoprotein receptor-related proteins 4, 5, and 6 to inhibit Wnt signaling. Since the initial discovery of sclerostin, much understanding has been gained into the role of this protein in the regulation of skeletal biology. In this article, we discuss the latest findings in the regulation of SOST expression, sclerostin mechanisms of action, and the potential utility of targeting sclerostin in conditions of low bone mass.


Subject(s)
Bone Density/physiology , Bone Morphogenetic Proteins/physiology , Genetic Markers/physiology , Wnt Signaling Pathway/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Gene Expression Regulation , Genetic Markers/genetics , Humans , LDL-Receptor Related Proteins , Signal Transduction/physiology
18.
Article in English | MEDLINE | ID: mdl-25599004

ABSTRACT

The adult skeleton undergoes bone remodeling that consists of bone formation by osteoblasts and bone resorption by osteoclasts. When the amount of bone resorbed is greater than the amount of new bone formed, low bone mass results, putting individuals at increased risk for osteoporosis and osteoporotic bone fracture. Nitrogenous bisphosphonates (NBPs) are the most common first line treatment for conditions of low bone mass. NBPs reduce osteoclast bone resorption by impairing the post-translational modification of small GTPases. Small GTPases play crucial roles in the differentiation, function, and survival of osteoclasts. Understanding the roles of individual small GTPases in osteoclast biology may lead to more targeted therapies for the treatment of low bone mass. In this review, we discuss recent investigations into the in vivo effects of individual GTPase deletion in osteoclasts and the molecular roles for small GTPases in osteoclast biology.

19.
Bone ; 57(1): 68-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23891907

ABSTRACT

The processes of bone resorption and bone formation are tightly coupled in young adults, which is crucial to maintenance of bone integrity. We have documented that osteoclasts secrete chemotactic agents to recruit osteoblast lineage cells, contributing to coupling. Bone formation subsequent to bone resorption becomes uncoupled with aging, resulting in significant bone loss. During bone resorption, osteoclasts release and activate transforming growth factor beta 1 (TGF-ß1) from the bone matrix; thus, elevated bone resorption increases the level of active TGF-ß in the local environment during aging. In this study, we examined the influences of TGF-ß1 on the ability of osteoclasts to recruit osteoblasts. TGF-ß1 increased osteoclast expression of the chemokine CXCL16 to promote osteoblast migration. TGF-ß1 also directly stimulated osteoblast migration; however, this direct response was blocked by conditioned medium from TGF-ß1-treated osteoclasts due to the presence of leukemia inhibitory factor (LIF) in the medium. CXCL16 and LIF expression was dependent on TGF-ß1 activation of Smad2 and Smad3. These results establish that TGF-ß1 induces CXCL16 and LIF production in osteoclasts, which modulate recruitment of osteoblasts to restore the bone lost during the resorptive phase of bone turnover.


Subject(s)
Chemokine CXCL6/metabolism , Leukemia Inhibitory Factor/metabolism , Osteoblasts/drug effects , Osteoclasts/drug effects , Transforming Growth Factor beta1/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CXCL16 , Chemokine CXCL6/pharmacology , Leukemia Inhibitory Factor/genetics , Mice , Osteoblasts/cytology , Osteoclasts/cytology
20.
Endocrinology ; 154(10): 3745-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23861379

ABSTRACT

In young adults, bone lost through osteoclast-mediated resorption is precisely replaced in both location and amount. Understanding how these two processes are coupled is crucial to advancing treatments for osteoporosis, a disease that progresses when the processes become uncoupled. We documented that osteoclasts secrete the mammalian integration 1 gene that is the homolog of Drosophila Wngless (Wnt) 10b, bone morphogenetic protein 6 (BMP6), and the chemokine sphingosin 1 phosphate (S1P) to promote mesenchymal cell mineralization in vitro. During bone resorption, TGF-ß1 is released from the bone extracellular matrix and activated by osteoclasts. Thus, TGF-ß1 levels are elevated during the resorption phase of bone turnover. We therefore investigated the influences of TGF-ß1 on osteoclast-mediated support of mineralization. TGF-ß1 increased osteoclast production of Wnt10b, but not BMP6 or S1P. Blocking Wnt10b activity with the Wnt signaling inhibitor Dickkoph-related protein 1 suppressed the ability of TGF-ß-treated osteoclast-conditioned media to promote osteoblast mineralization. Examination of TGF-ß signaling in osteoclasts revealed that induction of Wnt10b expression was dependent on Smad2/3 activation and independent from TGF-ß1 stimulation of protein kinase B (AKT) or MAPK kinase. TGF-ß1-treated osteoclast-conditioned media from cells with blocked Smad signaling exhibited a reduced ability to support mineralization, demonstrating the importance of Smad signaling in this response. Parallel cultures with suppressed TGF-ß activation of AKT or MAPK kinase signaling retained their ability to elevate mineralization. These results demonstrate that TGF-ß1 stimulates Wnt10b production in osteoclasts, which may enhance restoration of the bone lost during the resorptive phase of bone turnover.


Subject(s)
Calcification, Physiologic , Cell Communication , Osteoblasts/metabolism , Osteoclasts/metabolism , Transforming Growth Factor beta1/metabolism , Wnt Proteins/metabolism , Animals , Animals, Newborn , Bone Morphogenetic Protein 6/metabolism , Cells, Cultured , Culture Media, Conditioned/metabolism , Female , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoclasts/cytology , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...