Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37751740

ABSTRACT

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Subject(s)
Osteoclasts , RNA , Humans , Mice , Animals , Co-Repressor Proteins/genetics , Osteoclasts/metabolism , RANK Ligand/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Gene Expression
2.
Physiol Rev ; 97(1): 135-187, 2017 01.
Article in English | MEDLINE | ID: mdl-27807202

ABSTRACT

Estrogens and androgens influence the growth and maintenance of the mammalian skeleton and are responsible for its sexual dimorphism. Estrogen deficiency at menopause or loss of both estrogens and androgens in elderly men contribute to the development of osteoporosis, one of the most common and impactful metabolic diseases of old age. In the last 20 years, basic and clinical research advances, genetic insights from humans and rodents, and newer imaging technologies have changed considerably the landscape of our understanding of bone biology as well as the relationship between sex steroids and the physiology and pathophysiology of bone metabolism. Together with the appreciation of the side effects of estrogen-related therapies on breast cancer and cardiovascular diseases, these advances have also drastically altered the treatment of osteoporosis. In this article, we provide a comprehensive review of the molecular and cellular mechanisms of action of estrogens and androgens on bone, their influences on skeletal homeostasis during growth and adulthood, the pathogenetic mechanisms of the adverse effects of their deficiency on the female and male skeleton, as well as the role of natural and synthetic estrogenic or androgenic compounds in the pharmacotherapy of osteoporosis. We highlight latest advances on the crosstalk between hormonal and mechanical signals, the relevance of the antioxidant properties of estrogens and androgens, the difference of their cellular targets in different bone envelopes, the role of estrogen deficiency in male osteoporosis, and the contribution of estrogen or androgen deficiency to the monomorphic effects of aging on skeletal involution.


Subject(s)
Androgens/metabolism , Bone and Bones/metabolism , Bone and Bones/physiopathology , Estrogens/metabolism , Osteoporosis/physiopathology , Animals , Female , Homeostasis/physiology , Humans , Male , Osteoporosis/metabolism
4.
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
5.
J Biol Chem ; 289(35): 24069-78, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25002589

ABSTRACT

A decline of the levels and activity of Sirtuin1 (Sirt1), a NAD(+) class III histone deacetylase, with age contributes to the development of several diseases including type 2 diabetes, neurodegeneration, inflammation, and cancer. The anti-aging effects of Sirt1 evidently result from the deacetylation of many transcription factors and co-factors including members of the Forkhead box O (FoxO) family and ß-catenin. Wnt/ß-catenin is indispensable for osteoblast generation. FoxOs, on the other hand, sequester ß-catenin and inhibit osteoprogenitor proliferation. Here, we have deleted Sirt1 in osteoprogenitors expressing Osterix1 (Osx1)-Cre and their descendants. Sirt1(ΔOsx1) mice had lower cortical thickness in femora and vertebrae because of reduced bone formation at the endocortical surface. In line with this, osteoprogenitor cell cultures from the Sirt1(ΔOsx1) mice exhibited lower alkaline phosphatase activity and mineralization, as well as decreased proliferation and increased apoptosis. These changes were associated with decreased Wnt/ß-catenin signaling and expression of cyclin D1 and resulted from increased binding of FoxOs to ß-catenin. These findings demonstrate that Sirt1-induced deacetylation of FoxOs unleashes Wnt signaling. A decline in Sirt1 activity in osteoblast progenitors with aging may, therefore, contribute to the age-related loss of bone mass. Together with evidence that Sirt1 activators increase bone mass in aged mice, our results also suggest that Sirt1 could be a therapeutic target for osteoporosis.


Subject(s)
Bone Development/physiology , Forkhead Transcription Factors/physiology , Osteoblasts/metabolism , Sirtuin 1/physiology , Stem Cells/metabolism , beta Catenin/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Proliferation , DNA Primers , Gene Deletion , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirtuin 1/genetics , Wnt Proteins/metabolism
6.
J Biol Chem ; 288(42): 30064-30074, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-23990467

ABSTRACT

Iron is essential for osteoclast differentiation, and iron overload in a variety of hematologic diseases is associated with excessive bone resorption. Iron uptake by osteoclast precursors via the transferrin cycle increases mitochondrial biogenesis, reactive oxygen species production, and activation of cAMP response element-binding protein, a critical transcription factor downstream of receptor activator of NF-κB-ligand-induced calcium signaling. These changes are required for the differentiation of osteoclast precursors to mature bone-resorbing osteoclasts. However, the molecular mechanisms regulating cellular iron metabolism in osteoclasts remain largely unknown. In this report, we provide evidence that Steap4, a member of the six-transmembrane epithelial antigen of prostate (Steap) family proteins, is an endosomal ferrireductase with a critical role in cellular iron utilization in osteoclasts. Specifically, we show that Steap4 is the only Steap family protein that is up-regulated during osteoclast differentiation. Knocking down Steap4 expression in vitro by lentivirus-mediated short hairpin RNAs inhibits osteoclast formation and decreases cellular ferrous iron, reactive oxygen species, and the activation of cAMP response element-binding protein. These results demonstrate that Steap4 is a critical enzyme for cellular iron uptake and utilization in osteoclasts and, thus, indispensable for osteoclast development and function.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , FMN Reductase/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Osteoclasts/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Endosomes/genetics , Endosomes/metabolism , FMN Reductase/genetics , Gene Knockdown Techniques , Membrane Proteins/genetics , Mice , Osteoclasts/cytology
7.
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
8.
J Biol Chem ; 287(35): 29851-60, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22782898

ABSTRACT

Production of the cytokine receptor activator of NFκB ligand (RANKL) by lymphocytes has been proposed as a mechanism by which sex steroid deficiency causes bone loss. However, there have been no studies that functionally link RANKL expression in lymphocytes with bone loss in this condition. Herein, we examined whether RANKL expression in either B or T lymphocytes contributes to ovariectomy-induced bone loss in mice. Mice harboring a conditional RANKL allele were crossed with CD19-Cre or Lck-Cre mice to delete RANKL in B or T lymphocytes, respectively. Deletion of RANKL from either cell type had no impact on bone mass in estrogen-replete mice up to 7 months of age. However, mice lacking RANKL in B lymphocytes were partially protected from the bone loss caused by ovariectomy. This protection occurred in cancellous, but not cortical, bone and was associated with a failure to increase osteoclast numbers in the conditional knock-out mice. Deletion of RANKL from T lymphocytes had no impact on ovariectomy-induced bone loss. These results demonstrate that lymphocyte RANKL is not involved in basal bone remodeling, but B cell RANKL does contribute to the increase in osteoclasts and cancellous bone loss that occurs after loss of estrogen.


Subject(s)
B-Lymphocytes/metabolism , Bone Remodeling/immunology , Osteoclasts/metabolism , Osteoporosis/metabolism , RANK Ligand/metabolism , Alleles , Animals , B-Lymphocytes/immunology , Bone Density/genetics , Bone Density/immunology , Estrogens/genetics , Estrogens/immunology , Estrogens/metabolism , Female , Humans , Mice , Mice, Transgenic , Osteoclasts/immunology , Osteoporosis/genetics , Osteoporosis/immunology , Ovariectomy , RANK Ligand/genetics , RANK Ligand/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Bone ; 170: 116702, 2023 05.
Article in English | MEDLINE | ID: mdl-36773885

ABSTRACT

The scavenger receptor class B member 1 (SR-B1 or Scarb1) is a glycosylated cell surface receptor for high density lipoproteins (HDL), oxidized low density lipoproteins (OxLDL), and phosphocholine-containing oxidized phospholipids (PC-OxPLs). Scarb1 is expressed in macrophages and has been shown to have both pro- and anti-atherogenic properties. It has been reported that global deletion of Scarb1 in mice leads to either high or low bone mass and that PC-OxPLs decrease osteoblastogenesis and increase osteoclastogenesis. PC-OxPLs decrease bone mass in 6-month-old mice and are critical pathogenetic factors in the bone loss caused by high fat diet or aging. We have investigated here whether Scarb1 expression in myeloid cells affects bone mass and whether PC-OxPLs exert their anti-osteogenic effects via activation of Scarb1 in macrophages. To this end, we generated mice with deletion of Scarb1 in LysM-Cre expressing cells and found that lack of Scarb1 did not affect bone mass in vivo. These results indicate that Scarb1 expression in cells of the myeloid/osteoclast lineage does not contribute to bone homeostasis. Based on this evidence, and earlier studies of ours showing that Scarb1 expression in osteoblasts does not affect bone mass, we conclude that Scarb1 is not an important mediator of the adverse effects on PC-OxPLs in osteoclasts or osteoblasts in 6-month-old mice.


Subject(s)
Bone Density , Bone and Bones , Animals , Mice , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Bone and Bones/metabolism , Osteoclasts/metabolism , Osteogenesis
10.
J Biol Chem ; 286(52): 44326-35, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22030390

ABSTRACT

Endogenous glucocorticoids (GCs) and inflammatory cytokines contribute to the age-associated loss of bone mass and strength, but the molecular mechanisms responsible for their deleterious effects on the aging skeleton are unclear. Based on evidence that oxidative stress is a causal mechanism of the insulin resistance produced by either one of these two agents, we tested the hypothesis that their adverse skeletal effects also result from increased oxidative stress. We report that administration of prednisolone to mice increased reactive oxygen species (ROS) and the phosphorylation of p66(shc) (an amplifier of H(2)O(2) generation in mitochondria) in bone. Dexamethasone (Dex) and TNFα had a similar effect on osteoblastic cells in vitro. The generation of ROS by Dex and TNFα required PKCß/p66(shc) signaling and was responsible for the activation of JNK and induction of apoptosis by both agents. The activity of Forkhead box O (FoxO) transcription factors was also increased in response to ROS; however, FoxO activation opposed apoptosis induced by Dex and TNFα. In addition, both agents suppressed Akt phosphorylation as well as Wnt-induced proliferation and osteoblast differentiation. However, the inhibitory actions on Wnt signaling were independent of PKCß/p66(shc). Instead, they were mediated by inhibition of Akt and stimulation of FoxOs. These results demonstrate that ROS-induced activation of a PKCß/p66(shc)/JNK signaling cascade is responsible for the pro-apoptotic effects of Dex and TNFα on osteoblastic cells. Moreover, modulation of Akt and FoxOs by GCs and TNFα are cell-autonomous mechanisms of Wnt/ß-catenin antagonism contributing to the adverse effects of GC excess and inflammatory cytokines on bone alike.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Osteoblasts/metabolism , Oxidative Stress/drug effects , Prednisolone/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Wnt Signaling Pathway/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Knockout , Osteoblasts/cytology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins/genetics , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tumor Necrosis Factor-alpha/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/genetics , beta Catenin/metabolism
11.
N Engl J Med ; 360(1): 53-62, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118304

ABSTRACT

BACKGROUND: Bisphosphonates decrease bone resorption and are commonly used to treat or prevent osteoporosis. However, the effect of bisphosphonates on their target cells remains enigmatic, since in patients benefiting from therapy, little change, if any, has been observed in the number of osteoclasts, which are the cells responsible for bone resorption. METHODS: We examined 51 bone-biopsy specimens obtained after a 3-year, double-blind, randomized, placebo-controlled, dose-ranging trial of oral alendronate to prevent bone resorption among healthy postmenopausal women 40 through 59 years of age. The patients were assigned to one of five groups: those receiving placebo for 3 years; alendronate at a dose of 1, 5, or 10 mg per day for 3 years; or alendronate at a dose of 20 mg per day for 2 years, followed by placebo for 1 year. Formalin-fixed, undecalcified planar sections were assessed by bone histomorphometric methods. RESULTS: The number of osteoclasts was increased by a factor of 2.6 in patients receiving 10 mg of alendronate per day for 3 years as compared with the placebo group (P<0.01). Moreover, the number of osteoclasts increased as the cumulative dose of the drug increased (r=0.50, P<0.001). Twenty-seven percent of these osteoclasts were giant cells with pyknotic nuclei that were adjacent to superficial resorption cavities. Furthermore, giant, hypernucleated, detached osteoclasts with 20 to 40 nuclei were found after alendronate treatment had been discontinued for 1 year. Of these large cells, 20 to 37% were apoptotic, according to both their morphologic features and positive findings from in situ end labeling. CONCLUSIONS: Long-term alendronate treatment is associated with an increase in the number of osteoclasts, which include distinctive giant, hypernucleated, detached osteoclasts that are undergoing protracted apoptosis.


Subject(s)
Alendronate/pharmacology , Apoptosis/drug effects , Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Osteoclasts/drug effects , Acid Phosphatase/analysis , Adult , Alendronate/therapeutic use , Biopsy , Bone Density Conservation Agents/therapeutic use , Bone and Bones/cytology , Bone and Bones/pathology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Isoenzymes/analysis , Linear Models , Middle Aged , Osteoclasts/chemistry , Osteoclasts/cytology , Osteoclasts/pathology , Postmenopause , Tartrate-Resistant Acid Phosphatase
12.
PLoS One ; 17(3): e0265893, 2022.
Article in English | MEDLINE | ID: mdl-35349600

ABSTRACT

The scavenger receptor class B member 1 (SR-B1 or Scarb1) is a cell surface receptor for high density lipoproteins. It also binds oxidized low density lipoproteins and phosphocholine-containing oxidized phospholipids (PC-OxPL), which adversely affect bone homeostasis. Overexpression of a single chain form of the antigen-binding domain of E06 IgM-a natural antibody that recognizes PC-OxPL-increases trabecular and cortical bone mass in female and male mice by stimulating bone formation. We have previously reported that Scarb1 is the most abundant scavenger receptor for PC-OxPL in calvaria-derived osteoblastic cells. Additionally, bone marrow- and calvaria-derived osteoblasts from Scarb1 knockout mice (Scarb1 KO) are protected from the pro-apoptotic and anti-differentiating effects of OxPL. Previous skeletal analysis of Scarb1 KO mice has produced contradictory results, with some studies reporting elevated bone mass but another study reporting low bone mass. To clarify the role of Scarb1 in osteoblasts, we deleted Scarb1 specifically in cells of the osteoblast lineage using Osx1-Cre transgenic mice. We observed no difference in bone mineral density measured by DXA in either female or male Osx1-Cre;Scarb1fl/fl mice compared to wild type (WT), Osx1-Cre, or Scarb1fl/fl littermate controls. Additionally, microCT analysis of 6-month-old females and 7-month-old males did not detect any difference in trabecular or cortical bone mass between genotypes. These results indicate that expression of Scarb1 in cells of the osteoblast lineage does not play an important role in bone homeostasis and, therefore, it is not essential for the effects of PC-OxPL on these cells.


Subject(s)
Bone Density , Osteoblasts , Scavenger Receptors, Class B , Animals , Bone and Bones/diagnostic imaging , Female , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteogenesis , Receptors, Scavenger/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism
13.
Sci Rep ; 12(1): 10257, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715555

ABSTRACT

The protective effect of estrogens against cortical bone loss is mediated via direct actions on mesenchymal cells, but functional evidence for the mediators of these effects has only recently begun to emerge. We report that the matrix metalloproteinase 13 (MMP13) is the highest up-regulated gene in mesenchymal cells from mice lacking the estrogen receptor alpha (ERα). In sham-operated female mice with conditional Mmp13 deletion in Prrx1 expressing cells (Mmp13ΔPrrx1), the femur and tibia length was lower as compared to control littermates (Mmp13f./f). Additionally, in the sham-operated female Mmp13ΔPrrx1 mice cortical thickness and trabecular bone volume in the femur and tibia were higher and osteoclast number at the endocortical surfaces was lower, whereas bone formation rate was unaffected. Notably, the decrease of cortical thickness caused by ovariectomy (OVX) in the femur and tibia of Mmp13f./f mice was attenuated in the Mmp13ΔPrrx1 mice; but the decrease of trabecular bone caused by OVX was not affected. These results reveal that mesenchymal cell-derived MMP13 may regulate osteoclast number and/or activity, bone resorption, and bone mass. And increased production of mesenchymal cell-derived factors may be important mediators of the adverse effect of estrogen deficiency on cortical, but not trabecular, bone.


Subject(s)
Bone Density , Bone Diseases, Metabolic , Matrix Metalloproteinase 13/metabolism , Animals , Bone and Bones/diagnostic imaging , Cortical Bone , Estrogens , Female , Homeodomain Proteins , Humans , Matrix Metalloproteinase 13/genetics , Mice , Ovariectomy/adverse effects
14.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-33878033

ABSTRACT

Altered mitochondria activity in osteoblasts and osteoclasts has been implicated in the loss of bone mass associated with aging and estrogen deficiency - the 2 most common causes of osteoporosis. However, the mechanisms that control mitochondrial metabolism in bone cells during health or disease remain unknown. The mitochondrial deacetylase sirtuin-3 (Sirt3) has been earlier implicated in age-related diseases. Here, we show that deletion of Sirt3 had no effect on the skeleton of young mice but attenuated the age-related loss of bone mass in both sexes. This effect was associated with impaired bone resorption. Osteoclast progenitors from aged Sirt3-null mice were able to differentiate into osteoclasts, though the differentiated cells exhibited impaired polykaryon formation and resorptive activity, as well as decreased oxidative phosphorylation and mitophagy. The Sirt3 inhibitor LC-0296 recapitulated the effects of Sirt3 deletion in osteoclast formation and mitochondrial function, and its administration to aging mice increased bone mass. Deletion of Sirt3 also attenuated the increase in bone resorption and loss of bone mass caused by estrogen deficiency. These findings suggest that Sirt3 inhibition and the resulting impairment of osteoclast mitochondrial function could be a novel therapeutic intervention for the 2 most important causes of osteoporosis.


Subject(s)
Aging/physiology , Mitochondria/metabolism , Osteoporosis/metabolism , Sirtuin 3 , Animals , Estrogens/metabolism , Female , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism
15.
Aging Cell ; 20(8): e13442, 2021 08.
Article in English | MEDLINE | ID: mdl-34278710

ABSTRACT

Oxidized phospholipids (OxPLs) are pro-inflammatory molecules that affect bone remodeling under physiological conditions. Transgenic expression of a single-chain variable fragment (scFv) of the antigen-binding domain of E06, an IgM natural antibody that recognizes the phosphocholine (PC) moiety of OxPLs, increases trabecular and cortical bone in adult male and female mice by increasing bone formation. OxPLs increase with age, while natural antibodies decrease. Age-related bone loss is associated with increased oxidative stress and lipid peroxidation and is characterized by a decline in osteoblast number and bone formation, raising the possibility that increased OxPLs, together with the decline of natural antibodies, contribute to age-related bone loss. We show here that transgenic expression of E06-scFv attenuated the age-associated loss of spinal, femoral, and total bone mineral density in both female and male mice aged up to 22 and 24 months, respectively. E06-scFv attenuated the age-associated decline in trabecular bone, but not cortical bone, and this effect was associated with an increase in osteoblasts and a decrease in osteoclasts. Furthermore, RNA-seq analysis showed that E06-scFv increased Wnt10b expression in vertebral bone in aged mice, indicating that blocking OxPLs increases Wnt signaling. Unlike age-related bone loss, E06-scFv did not attenuate the bone loss caused by estrogen deficiency or unloading in adult mice. These results demonstrate that OxPLs contribute to age-associated bone loss. Neutralization of OxPLs, therefore, is a promising therapeutic target for senile osteoporosis, as well as atherosclerosis and non-alcoholic steatohepatitis (NASH), two other conditions shown to be attenuated by E06-scFv in mice.


Subject(s)
Aging/pathology , Bone Diseases, Metabolic/physiopathology , Phospholipids/metabolism , Animals , Female , Male , Mice , Mice, Transgenic
16.
J Bone Miner Res ; 36(1): 170-185, 2021 01.
Article in English | MEDLINE | ID: mdl-32990984

ABSTRACT

Oxidized phospholipids containing phosphocholine (OxPL) are pro-inflammatory lipid peroxidation products that bind to scavenger receptors (SRs), such as Scarb1, and toll-like receptors (TLRs). Excessive OxPL, as found in oxidized low-density lipoprotein (OxLDL), overwhelm these defense mechanisms and become pathogenic in atherosclerosis, nonalcoholic steatohepatitis (NASH), and osteoporosis. We previously reported that the innate IgM natural antibody E06 binds to OxPL and neutralizes their deleterious effects; expression of the single-chain (scFv) form of the antigen-binding domain of E06 (E06-scFv) as a transgene increases trabecular bone in male mice. We show herein that E06-scFv increases trabecular and cortical bone in female and male mice by increasing bone formation and decreasing osteoblast apoptosis in vivo. Homozygous E06-scFv mice have higher bone mass than hemizygous, showing a dose effect of the transgene. To investigate how OxPL restrain bone formation under physiologic conditions, we measured the levels of SRs and TLRs that bind OxPL. We found that osteoblastic cells primarily express Scarb1. Moreover, OxLDL-induced apoptosis and reduced differentiation were prevented in bone marrow-derived or calvaria-derived osteoblasts from Scarb1 knockout mice. Because Scarb1-deficient mice are reported to have high bone mass, our results suggest that E06 may promote bone anabolism in healthy young mice, at least in part, by neutralizing OxPL, which in turn promote Scarb1-mediated apoptosis of osteoblasts or osteoblast precursors. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)..


Subject(s)
Osteogenesis , Phospholipids , Animals , Antibodies, Neutralizing , Female , Male , Mice , Mice, Knockout , Oxidation-Reduction
17.
J Biol Chem ; 284(40): 27438-48, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19657144

ABSTRACT

Loss of bone mass with advancing age in mice is because of decreased osteoblast number and is associated with increased oxidative stress and decreased canonical Wnt signaling. However, the underlying mechanisms are poorly understood. We report an age-related increase in the lipid oxidation product 4-hydroxynonenal (4-HNE) as well as increased expression of lipoxygenase and peroxisome proliferator-activated receptor-gamma (PPARgamma) in the murine skeleton. These changes together with decreased Wnt signaling are reproduced in 4-month-old mice bearing a high expressing allele of the lipoxygenase Alox15. The addition of 4-HNE to cultured osteoblastic cells increases oxidative stress, which in turn diverts beta-catenin from T-cell-specific transcription factors to Forkhead box O (FoxO) transcription factors, thereby attenuating the suppressive effect of beta-catenin on PPARgamma gene expression. Oxidized lipids, acting as ligands of PPARgamma, promote binding of PPARgamma2 to beta-catenin and reduce the levels of the latter, and they attenuate Wnt3a-stimulated proliferation and osteoblast differentiation. Furthermore, oxidized lipids and 4-HNE stimulate apoptosis of osteoblastic cells. In view of the role of oxidized lipids in atherogenesis, the adverse effects of lipoxygenase-mediated lipid oxidation on the differentiation and survival of osteoblasts may provide a mechanistic explanation for the link between atherosclerosis and osteoporosis.


Subject(s)
Gene Expression Regulation , Lipid Metabolism , Osteogenesis , Oxidative Stress , PPAR gamma/metabolism , Signal Transduction , Wnt Proteins/metabolism , Aging/metabolism , Aging/physiology , Aldehydes/pharmacology , Alleles , Animals , Apoptosis , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Bone and Bones/cytology , Bone and Bones/drug effects , Bone and Bones/metabolism , Cattle , Cell Count , Cell Line , Cell Survival , Female , Gene Expression Regulation, Enzymologic , Male , Mice , Multienzyme Complexes/genetics , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , PPAR gamma/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects
18.
Mol Cell Biol ; 27(4): 1516-30, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17158928

ABSTRACT

Estrogens control gene transcription by cis or trans interactions of the estrogen receptor (ER) with target DNA or via the activation of cytoplasmic kinases. We report that selective activation of kinase-mediated actions of the ER with 4-estren-3alpha,17beta-diol (estren) or an estradiol-dendrimer conjugate, each a synthetic compound that stimulates kinase-mediated ER actions 1,000 to 10,000 times more potently than direct DNA interactions, induced osteoblastic differentiation in established cell lines of uncommitted osteoblast precursors and primary cultures of osteoblast progenitors by stimulating Wnt and BMP-2 signaling in a kinase-dependent manner. In sharp contrast, 17beta-estradiol (E(2)) suppressed BMP-2-induced osteoblast progenitor commitment and differentiation. Consistent with the in vitro findings, estren, but not E(2), stimulated Wnt/beta-catenin-mediated transcription in T-cell factor-lacZ transgenic mice. Moreover, E(2) stimulated BMP signaling in mice in which ERalpha lacks DNA binding activity and classical estrogen response element-mediated transcription (ERalpha(NERKI/-)) but not in wild-type controls. This evidence reveals for the first time the existence of a large signalosome in which inputs from the ER, kinases, bone morphogenetic proteins, and Wnt signaling converge to induce differentiation of osteoblast precursors. ER can either induce it or repress it, depending on whether the activating ligand (and presumably the resulting conformation of the receptor protein) precludes or accommodates ERE-mediated transcription.


Subject(s)
Cell Differentiation , Extracellular Signal-Regulated MAP Kinases/metabolism , Osteoblasts/cytology , Osteoblasts/enzymology , Receptors, Estrogen/metabolism , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Dihydrotestosterone/pharmacology , Enzyme Activation/drug effects , Estradiol/pharmacology , Estrenes/pharmacology , Female , Humans , Male , Mice , Osteoblasts/drug effects , Phosphorylation/drug effects , Receptors, Androgen/metabolism , Response Elements/drug effects , Signal Transduction/drug effects , Smad Proteins/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects , Wnt Proteins/genetics , Wnt Proteins/metabolism
19.
J Bone Miner Res ; 35(8): 1441-1451, 2020 08.
Article in English | MEDLINE | ID: mdl-32154948

ABSTRACT

CXCL12 is abundantly expressed in reticular cells associated with the perivascular niches of the bone marrow (BM) and is indispensable for B lymphopoiesis. Cxcl12 promotes osteoclastogenesis and has been implicated in pathologic bone resorption. We had shown earlier that estrogen receptor α deletion in osteoprogenitors and estrogen deficiency in mice increase Cxcl12 mRNA and protein levels in the BM plasma, respectively. We have now generated female and male mice with conditional deletion of a Cxcl12 allele in Prrx1 targeted cells (Cxcl12∆Prrx1 ) and show herein that they have a 90% decrease in B lymphocytes but increased erythrocytes and adipocytes in the marrow. Ovariectomy increased the expression of Cxcl12 and B-cell number in the Cxcl12f/f control mice, but these effects were abrogated in the Cxcl12∆Prrx1 mice. Cortical bone mass was not affected in Cxcl12∆Prrx1 mice. Albeit, the cortical bone loss caused by ovariectomy was greatly attenuated. Most unexpectedly, the rate of bone turnover in sex steroid-sufficient female or male Cxcl12∆Prrx1 mice was dramatically increased, as evidenced by a more than twofold increase in several osteoblast- and osteoclast-specific mRNAs, as well as increased mineral apposition and bone formation rate and increased osteoclast number in the endosteal surface. The magnitude of the Cxcl12∆Prrx1 -induced changes were much greater than those caused by ovariectomy or orchidectomy in the Cxcl12f/f mice. These results strengthen the evidence that CXCL12 contributes to the loss of cortical bone mass caused by estrogen deficiency. Moreover, they reveal for the first time that in addition to its effects on hematopoiesis, CXCL12 restrains bone turnover-without changing the balance between resorption and formation-by suppressing osteoblastogenesis and the osteoclastogenesis support provided by cells of the osteoblast lineage. © 2020 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption , Bone and Bones , Chemokine CXCL12 , Estrogens/deficiency , Animals , Bone Remodeling , Bone Resorption/genetics , Chemokine CXCL12/genetics , Cortical Bone/diagnostic imaging , Female , Gene Deletion , Homeodomain Proteins , Male , Mice , Osteoblasts , Osteoclasts , Ovariectomy
20.
Aging Cell ; 19(11): e13247, 2020 11.
Article in English | MEDLINE | ID: mdl-33048436

ABSTRACT

Marrow adipocytes and osteoblasts differentiate from common mesenchymal progenitors in a mutually exclusive manner, and diversion of these progenitors toward adipocytes in old age has been proposed to account for the decline in osteoblasts and the development of involutional osteoporosis. This idea has been supported by evidence that thiazolidinedione (TZD)-induced activation of PPARγ, the transcription factor required for adipocyte differentiation, increases marrow fat and causes bone loss. We functionally tested this hypothesis using C57BL/6J mice with conditional deletion of PPARγ from early mesenchymal progenitors targeted by the Prx1-Cre transgene. Using a longitudinal littermate-controlled study design, we observed that PPARγ is indispensable for TZD-induced increase in marrow adipocytes in 6-month-old male mice, and age-associated increase in marrow adipocytes in 22-month-old female mice. In contrast, PPARγ is dispensable for the loss of cortical and trabecular bone caused by TZD or old age. Instead, PPARγ restrains age-dependent development of cortical porosity. These findings do not support the long-standing hypothesis that increased marrow adipocyte differentiation contributes to bone loss in old age but reveal a novel role of mesenchymal cell PPARγ in the maintenance of cortical integrity.


Subject(s)
Adipogenesis/physiology , Osteoporosis/physiopathology , Age Factors , Animals , Cell Differentiation , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL