Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(5): 1137-1139, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636131

ABSTRACT

In this issue of Cell, McDonald et al. show that giant multinucleated, bone-resorbing osteoclasts dissolve into smaller cells, termed "osteopmorhs," which re-form into osteoclasts at distal bone sites (McDonald et al., 2021). These findings overturn the long-standing premise that osteoclasts differentiate solely from hematopoietic precursors and undergo apoptosis after completing resorption.


Subject(s)
Bone Resorption , Bone and Bones , Humans , Osteoclasts
2.
Nature ; 603(7901): 470-476, 2022 03.
Article in English | MEDLINE | ID: mdl-35236988

ABSTRACT

Alzheimer's disease has a higher incidence in older women, with a spike in cognitive decline that tracks with visceral adiposity, dysregulated energy homeostasis and bone loss during the menopausal transition1,2. Inhibiting the action of follicle-stimulating hormone (FSH) reduces body fat, enhances thermogenesis, increases bone mass and lowers serum cholesterol in mice3-7. Here we show that FSH acts directly on hippocampal and cortical neurons to accelerate amyloid-ß and Tau deposition and impair cognition in mice displaying features of Alzheimer's disease. Blocking FSH action in these mice abrogates the Alzheimer's disease-like phenotype by inhibiting the neuronal C/EBPß-δ-secretase pathway. These data not only suggest a causal role for rising serum FSH levels in the exaggerated Alzheimer's disease pathophysiology during menopause, but also reveal an opportunity for treating Alzheimer's disease, obesity, osteoporosis and dyslipidaemia with a single FSH-blocking agent.


Subject(s)
Alzheimer Disease , Follicle Stimulating Hormone , Aged , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Bone Density , Cognition , Female , Follicle Stimulating Hormone/metabolism , Humans , Mice , Thermogenesis
3.
Nature ; 546(7656): 107-112, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538730

ABSTRACT

Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the ß-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the ß-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.


Subject(s)
Adipose Tissue/metabolism , Adiposity , Follicle Stimulating Hormone, beta Subunit/antagonists & inhibitors , Thermogenesis , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Antibodies/immunology , Antibodies/pharmacology , Diet, High-Fat/adverse effects , Female , Follicle Stimulating Hormone, beta Subunit/immunology , Haploinsufficiency , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/drug therapy , Obesity/prevention & control , Osteoporosis/drug therapy , Ovariectomy , Oxygen Consumption/drug effects , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/genetics , Receptors, FSH/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1/biosynthesis
4.
Proc Natl Acad Sci U S A ; 117(25): 14386-14394, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513693

ABSTRACT

We report that two widely-used drugs for erectile dysfunction, tadalafil and vardenafil, trigger bone gain in mice through a combination of anabolic and antiresorptive actions on the skeleton. Both drugs were found to enhance osteoblastic bone formation in vivo using a unique gene footprint and to inhibit osteoclast formation. The target enzyme, phosphodiesterase 5A (PDE5A), was found to be expressed in mouse and human bone as well as in specific brain regions, namely the locus coeruleus, raphe pallidus, and paraventricular nucleus of the hypothalamus. Localization of PDE5A in sympathetic neurons was confirmed by coimmunolabeling with dopamine ß-hydroxylase, as well as by retrograde bone-brain tracing using a sympathetic nerve-specific pseudorabies virus, PRV152. Both drugs elicited an antianabolic sympathetic imprint in osteoblasts, but with net bone gain. Unlike in humans, in whom vardenafil is more potent than tadalafil, the relative potencies were reversed with respect to their osteoprotective actions in mice. Structural modeling revealed a higher binding energy of tadalafil to mouse PDE5A compared with vardenafil, due to steric clashes of vardenafil with a single methionine residue at position 806 in mouse PDE5A. Collectively, our findings suggest that a balance between peripheral and central actions of PDE5A inhibitors on bone formation together with their antiresorptive actions specify the osteoprotective action of PDE5A blockade.


Subject(s)
Erectile Dysfunction/drug therapy , Osteogenesis/drug effects , Osteoporosis/drug therapy , Phosphodiesterase 5 Inhibitors/pharmacology , Aging/physiology , Animals , Bone Density/drug effects , Bone Density/physiology , Bone and Bones/cytology , Bone and Bones/drug effects , Bone and Bones/metabolism , Brain/cytology , Brain/drug effects , Brain/metabolism , Cell Differentiation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Drug Repositioning , Erectile Dysfunction/complications , Humans , Male , Mice , Middle Aged , Models, Animal , Models, Molecular , Neurons/drug effects , Neurons/metabolism , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/physiology , Osteoporosis/complications , Osteoporotic Fractures/etiology , Osteoporotic Fractures/prevention & control , Phosphodiesterase 5 Inhibitors/chemistry , Phosphodiesterase 5 Inhibitors/therapeutic use , Primary Cell Culture , Tadalafil/chemistry , Tadalafil/pharmacology , Tadalafil/therapeutic use , Vardenafil Dihydrochloride/chemistry , Vardenafil Dihydrochloride/pharmacology , Vardenafil Dihydrochloride/therapeutic use
5.
Proc Natl Acad Sci U S A ; 117(46): 28971-28979, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33127753

ABSTRACT

Blocking the action of FSH genetically or pharmacologically in mice reduces body fat, lowers serum cholesterol, and increases bone mass, making an anti-FSH agent a potential therapeutic for three global epidemics: obesity, osteoporosis, and hypercholesterolemia. Here, we report the generation, structure, and function of a first-in-class, fully humanized, epitope-specific FSH blocking antibody with a KD of 7 nM. Protein thermal shift, molecular dynamics, and fine mapping of the FSH-FSH receptor interface confirm stable binding of the Fab domain to two of five receptor-interacting residues of the FSHß subunit, which is sufficient to block its interaction with the FSH receptor. In doing so, the humanized antibody profoundly inhibited FSH action in cell-based assays, a prelude to further preclinical and clinical testing.


Subject(s)
Adipose Tissue/metabolism , Antibodies, Blocking/immunology , Bone and Bones/metabolism , Epitopes , Follicle Stimulating Hormone/immunology , Animals , Antibodies, Blocking/chemistry , Antibodies, Monoclonal , Bone Density , Female , Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone, beta Subunit/immunology , Humans , Hypercholesterolemia , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Obesity , Osteoporosis , Receptors, FSH/metabolism
6.
Proc Natl Acad Sci U S A ; 116(52): 26808-26815, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31843930

ABSTRACT

The primitive neurohypophyseal nonapeptide oxytocin (OXT) has established functions in parturition, lactation, appetite, and social behavior. We have shown that OXT has direct actions on the mammalian skeleton, stimulating bone formation by osteoblasts and modulating the genesis and function of bone-resorbing osteoclasts. We deleted OXT receptors (OXTRs) selectively in osteoblasts and osteoclasts using Col2.3Cre and Acp5Cre mice, respectively. Both male and female Col2.3Cre+:Oxtrfl/fl mice recapitulate the low-bone mass phenotype of Oxtr+/- mice, suggesting that OXT has a prominent osteoblastic action in vivo. Furthermore, abolishment of the anabolic effect of estrogen in Col2.3Cre+:Oxtrfl/fl mice suggests that osteoblastic OXTRs are necessary for estrogen action. In addition, the high bone mass in Acp5Cre+:Oxtrfl/fl mice indicates a prominent action of OXT in stimulating osteoclastogenesis. In contrast, we found that in pregnant and lactating Col2.3Cre+:Oxtrfl/fl mice, elevated OXT inhibits bone resorption and rescues the bone loss otherwise noted during pregnancy and lactation. However, OXT does not contribute to ovariectomy-induced bone loss. Finally, we show that OXT acts directly on OXTRs on adipocytes to suppress the white-to-beige transition gene program. Despite this direct antibeiging action, injected OXT reduces total body fat, likely through an action on OXT-ergic neurons. Consistent with an antiobesity action of OXT, Oxt-/- and Oxtr-/- mice display increased total body fat. Overall, the actions of OXT on bone mass and body composition provide the framework for future therapies for osteoporosis and obesity.

7.
Proc Natl Acad Sci U S A ; 115(9): 2192-2197, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440419

ABSTRACT

Pituitary hormones have long been thought solely to regulate single targets. Challenging this paradigm, we discovered that both anterior and posterior pituitary hormones, including FSH, had other functions in physiology. We have shown that FSH regulates skeletal integrity, and, more recently, find that FSH inhibition reduces body fat and induces thermogenic adipose tissue. A polyclonal antibody raised against a short, receptor-binding epitope of FSHß was found not only to rescue bone loss postovariectomy, but also to display marked antiobesity and probeiging actions. Questioning whether a single agent could be used to treat two medical conditions of public health importance--osteoporosis and obesity--we developed two further monoclonal antibodies, Hf2 and Mf4, against computationally defined receptor-binding epitopes of FSHß. Hf2 has already been shown to reduce body weight and fat mass and cause beiging in mice on a high-fat diet. Here, we show that Hf2, which binds mouse Fsh in immunoprecipitation assays, also increases cortical thickness and trabecular bone volume, and microstructural parameters, in sham-operated and ovariectomized mice, noted on microcomputed tomography. This effect was largely recapitulated with Mf4, which inhibited bone resorption by osteoclasts and stimulated new bone formation by osteoblasts. These effects were exerted in the absence of alterations in serum estrogen in wild-type mice. We also reconfirm the existence of Fshrs in bone by documenting the specific binding of fluorescently labeled FSH, FSH-CH, in vivo. Our study provides the framework for the future development of an FSH-based therapeutic that could potentially target both bone and fat.


Subject(s)
Antibodies, Monoclonal/pharmacology , Epitopes , Follicle Stimulating Hormone, beta Subunit/immunology , Animals , Antibody Specificity , Bone Density , Bone Resorption , Catalytic Domain , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Ovariectomy , Protein Binding , Protein Conformation
8.
FASEB J ; 33(8): 9167-9181, 2019 08.
Article in English | MEDLINE | ID: mdl-31063702

ABSTRACT

The mitochondria-to-nucleus retrograde signaling (MtRS) pathway aids in cellular adaptation to stress. We earlier reported that the Ca2+- and calcineurin-dependent MtRS induces macrophage differentiation to bone-resorbing osteoclasts. However, mechanisms through which macrophages sense and respond to cellular stress remain unclear. Here, we induced mitochondrial stress in macrophages by knockdown (KD) of subunits IVi1 or Vb of cytochrome c oxidase (CcO). Whereas both IVi1 and Vb KD impair CcO activity, IVi1 KD cells produced higher levels of cellular and mitochondrial reactive oxygen species with increased glycolysis. Additionally, IVi1 KD induced the activation of MtRS factors NF-κB, NFAT2, and C/EBPδ as well as inflammatory cytokines, NOS 2, increased phagocytic activity, and a greater osteoclast differentiation potential at suboptimal RANK-L concentrations. The osteoclastogenesis in IVi1 KD cells was reversed fully with an IL-6 inhibitor LMT-28, whereas there was minimal rescue of the enhanced phagocytosis in these cells. In agreement with our findings in cultured macrophages, primary bone marrow-derived macrophages from MPV17-/- mice, a model for mitochondrial dysfunction, also showed higher propensity for osteoclast formation. This is the first report showing that CcO dysfunction affects inflammatory pathways, phagocytic function, and osteoclastogenesis.-Angireddy, R., Kazmi, H. R., Srinivasan, S., Sun, L., Iqbal, J., Fuchs, S. Y., Guha, M., Kijima, T., Yuen, T., Zaidi, M., Avadhani, N. G. Cytochrome c oxidase dysfunction enhances phagocytic function and osteoclast formation in macrophages.


Subject(s)
Electron Transport Complex IV/metabolism , Macrophages/cytology , Macrophages/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Phagocytosis/physiology , Animals , Cell Differentiation , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Gene Knockdown Techniques , Macrophages/classification , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mitochondria/metabolism , Osteogenesis , RAW 264.7 Cells , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Stress, Physiological
9.
Proc Natl Acad Sci U S A ; 111(50): 17995-8000, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25453078

ABSTRACT

A variety of human cancers, including nonsmall cell lung (NSCLC), breast, and colon cancers, are driven by the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. Having shown that bisphosphonates, a class of drugs used widely for the therapy of osteoporosis and metastatic bone disease, reduce cancer cell viability by targeting HER1, we explored their potential utility in the prevention and therapy of HER-driven cancers. We show that bisphosphonates inhibit colony formation by HER1(ΔE746-A750)-driven HCC827 NSCLCs and HER1(wt)-expressing MB231 triple negative breast cancers, but not by HER(low)-SW620 colon cancers. In parallel, oral gavage with bisphosphonates of mice xenografted with HCC827 or MB231 cells led to a significant reduction in tumor volume in both treatment and prevention protocols. This result was not seen with mice harboring HER(low) SW620 xenografts. We next explored whether bisphosphonates can serve as adjunctive therapies to tyrosine kinase inhibitors (TKIs), namely gefitinib and erlotinib, and whether the drugs can target TKI-resistant NSCLCs. In silico docking, together with molecular dynamics and anisotropic network modeling, showed that bisphosphonates bind to TKIs within the HER1 kinase domain. As predicted from this combinatorial binding, bisphosphonates enhanced the effects of TKIs in reducing cell viability and driving tumor regression in mice. Impressively, the drugs also overcame erlotinib resistance acquired through the gatekeeper mutation T790M, thus offering an option for TKI-resistant NSCLCs. We suggest that bisphosphonates can potentially be repurposed for the prevention and adjunctive therapy of HER1-driven cancers.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/prevention & control , Diphosphonates/pharmacology , ErbB Receptors/antagonists & inhibitors , Animals , Blotting, Western , Diphosphonates/therapeutic use , Drug Repositioning/methods , Female , Flow Cytometry , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Molecular Dynamics Simulation , Protein Binding , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles , Tumor Stem Cell Assay
10.
Proc Natl Acad Sci U S A ; 111(50): 17989-94, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25453081

ABSTRACT

Bisphosphonates are the most commonly prescribed medicines for osteoporosis and skeletal metastases. The drugs have also been shown to reduce cancer progression, but only in certain patient subgroups, suggesting that there is a molecular entity that mediates bisphosphonate action on tumor cells. Using connectivity mapping, we identified human epidermal growth factor receptors (human EGFR or HER) as a potential new molecular entity for bisphosphonate action. Protein thermal shift and cell-free kinase assays, together with computational modeling, demonstrated that N-containing bisphosphonates directly bind to the kinase domain of HER1/2 to cause a global reduction in downstream signaling. By doing so, the drugs kill lung, breast, and colon cancer cells that are driven by activating mutations or overexpression of HER1. Knocking down HER isoforms thus abrogates cell killing by bisphosphonates, establishing complete HER dependence and ruling out a significant role for other receptor tyrosine kinases or the enzyme farnesyl pyrophosphate synthase. Consistent with this finding, colon cancer cells expressing low levels of HER do not respond to bisphosphonates. The results suggest that bisphosphonates can potentially be repurposed for the prevention and therapy of HER family-driven cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Diphosphonates/pharmacology , ErbB Receptors/antagonists & inhibitors , Models, Molecular , Anisotropy , Blotting, Western , Cell Line, Tumor , Crystallography , Diphosphonates/metabolism , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Fluorescence , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Tetrazolium Salts , Thiazoles
11.
Proc Natl Acad Sci U S A ; 110(27): 11115-20, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776235

ABSTRACT

Smoking is a major risk factor for osteoporosis and fracture, but the mechanism through which smoke causes bone loss remains unclear. Here, we show that the smoke toxins benzo(a)pyrene (BaP) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) interact with the aryl hydrocarbon receptor (Ahr) to induce osteoclastic bone resorption through the activation of cytochrome P450 1a/1b (Cyp1) enzymes. BaP and TCDD enhanced osteoclast formation in bone marrow cell cultures and gavage with BaP stimulated bone resorption and osteoclastogenesis in vivo. The osteoclastogenesis triggered by BaP or RANK-L was reduced in Ahr(-/-) cells, consistent with the high bone mass noted in Ahr(-/-) male mice. The receptor activator of NF-κB ligand (RANK-L) also failed to induce the expression of Cyp1 enzymes in Ahr(-/-) cells. Furthermore, the osteoclastogenesis induced by TCDD was lower in Cyp1a1/1a2(-/-) and Cyp1a1/1a2/1b1(-/-) cultures, indicating that Ahr was upstream of the Cyp enzymes. Likewise, the pharmacological inhibition of the Cyp1 enzymes with tetramethylsilane or proadifen reduced osteoclastogenesis. Finally, deletion of the Cyp1a1, Cyp1a2, and Cyp1b1 in triple knockout mice resulted in reduced bone resorption and recapitulated the high bone mass phenotype of Ahr(-/-) mice. Overall, the data identify the Ahr and Cyp1 enzymes not only in the pathophysiology of smoke-induced osteoporosis, but also as potential targets for selective modulation by new therapeutics.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Bone Resorption/etiology , Bone Resorption/metabolism , Carcinogens/toxicity , Receptors, Aryl Hydrocarbon/physiology , Smoke/adverse effects , Animals , Aryl Hydrocarbon Hydroxylases/deficiency , Aryl Hydrocarbon Hydroxylases/genetics , Benzo(a)pyrene/toxicity , Bone Resorption/pathology , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/deficiency , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1A2/deficiency , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1B1 , Disease Models, Animal , Enzyme Induction/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Smoking/adverse effects , Smoking/genetics , Nicotiana/toxicity
12.
Proc Natl Acad Sci U S A ; 109(25): 10018-23, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22665763

ABSTRACT

Inherited deficiency of acid ß-glucosidase (GCase) due to biallelic mutations in the GBA (glucosidase, ß, acid) gene causes the classic manifestations of Gaucher disease (GD) involving the viscera, the skeleton, and the lungs. Clinical observations point to immune defects in GD beyond the accumulation of activated macrophages engorged with lysosomal glucosylceramide. Here, we show a plethora of immune cell aberrations in mice in which the GBA gene is deleted conditionally in hematopoietic stem cells (HSCs). The thymus exhibited the earliest and most striking alterations reminiscent of impaired T-cell maturation, aberrant B-cell recruitment, enhanced antigen presentation, and impaired egress of mature thymocytes. These changes correlated strongly with disease severity. In contrast to the profound defects in the thymus, there were only limited cellular defects in peripheral lymphoid organs, mainly restricted to mice with severe disease. The cellular changes in GCase deficiency were accompanied by elevated T-helper (Th)1 and Th2 cytokines that also tracked with disease severity. Finally, the proliferation of GCase-deficient HSCs was inhibited significantly by both GL1 and Lyso-GL1, suggesting that the "supply" of early thymic progenitors from bone marrow may, in fact, be reduced in GBA deficiency. The results not only point to a fundamental role for GBA in immune regulation but also suggest that GBA mutations in GD may cause widespread immune dysregulation through the accumulation of substrates.


Subject(s)
Gaucher Disease/immunology , Glucosylceramidase/genetics , Animals , Antigens, CD/immunology , Gaucher Disease/genetics , Immunophenotyping , Mice , Mice, Knockout
13.
J Biol Chem ; 287(34): 29159-67, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22761429

ABSTRACT

Estrogen uses two mechanisms to exert its effect on the skeleton: it inhibits bone resorption by osteoclasts and, at higher doses, can stimulate bone formation. Although the antiresorptive action of estrogen arises from the inhibition of the MAPK JNK, the mechanism of its effect on the osteoblast remains unclear. Here, we report that the anabolic action of estrogen in mice occurs, at least in part, through oxytocin (OT) produced by osteoblasts in bone marrow. We show that the absence of OT receptors (OTRs) in OTR(-/-) osteoblasts or attenuation of OTR expression in silenced cells inhibits estrogen-induced osteoblast differentiation, transcription factor up-regulation, and/or OT production in vitro. In vivo, OTR(-/-) mice, known to have a bone formation defect, fail to display increases in trabecular bone volume, cortical thickness, and bone formation in response to estrogen. Furthermore, osteoblast-specific Col2.3-Cre(+)/OTR(fl/fl) mice, but not TRAP-Cre(+)/OTR(fl/fl) mice, mimic the OTR(-/-) phenotype and also fail to respond to estrogen. These data attribute the phenotype of OTR deficiency to an osteoblastic rather than an osteoclastic defect. Physiologically, feed-forward OT release in bone marrow by a rising estrogen concentration may facilitate rapid skeletal recovery during the latter phases of lactation.


Subject(s)
Bone and Bones/metabolism , Estrogens/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , Osteogenesis/physiology , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Animals , Bone and Bones/cytology , Female , JNK Mitogen-Activated Protein Kinases/genetics , Lactation/physiology , Mice , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Oxytocin/genetics , Receptors, Oxytocin/genetics
15.
Proc Natl Acad Sci U S A ; 107(19): 8782-7, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20421485

ABSTRACT

We report that adrenocorticotropic hormone (ACTH) protects against osteonecrosis of the femoral head induced by depot methylprednisolone acetate (depomedrol). This therapeutic response likely arises from enhanced osteoblastic support and the stimulation of VEGF by ACTH; the latter is largely responsible for maintaining the fine vascular network that surrounds highly remodeling bone. We suggest examining the efficacy of ACTH in preventing human osteonecrosis, a devastating complication of glucocorticoid therapy.


Subject(s)
Adrenocorticotropic Hormone/therapeutic use , Femur/pathology , Glucocorticoids/adverse effects , Osteonecrosis/chemically induced , Osteonecrosis/drug therapy , Protective Agents/therapeutic use , 3T3 Cells , Adrenocorticotropic Hormone/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cytoprotection/drug effects , Female , Femur/drug effects , Humans , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteonecrosis/prevention & control , Protective Agents/pharmacology , Rabbits , Vascular Endothelial Growth Factor A/biosynthesis
16.
Proc Natl Acad Sci U S A ; 107(45): 19473-8, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20962279

ABSTRACT

In nonneuronopathic type 1 Gaucher disease (GD1), mutations in the glucocerebrosidase gene (GBA1) gene result in glucocerebrosidase deficiency and the accumulation of its substrate, glucocerebroside (GL-1), in the lysosomes of mononuclear phagocytes. This prevailing macrophage-centric view, however, does not explain emerging aspects of the disease, including malignancy, autoimmune disease, Parkinson disease, and osteoporosis. We conditionally deleted the GBA1 gene in hematopoietic and mesenchymal cell lineages using an Mx1 promoter. Although this mouse fully recapitulated human GD1, cytokine measurements, microarray analysis, and cellular immunophenotyping together revealed widespread dysfunction not only of macrophages, but also of thymic T cells, dendritic cells, and osteoblasts. The severe osteoporosis was caused by a defect in osteoblastic bone formation arising from an inhibitory effect of the accumulated lipids LysoGL-1 and GL-1 on protein kinase C. This study provides direct evidence for the involvement in GD1 of multiple cell lineages, suggesting that cells other than macrophages may be worthwhile therapeutic targets.


Subject(s)
Gaucher Disease/pathology , Gene Deletion , Glucosylceramidase/deficiency , Macrophages/pathology , Animals , Gaucher Disease/genetics , Glucosylceramidase/genetics , Hematopoietic Stem Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Mice , Osteoporosis/etiology , Phenotype , Promoter Regions, Genetic
17.
Biochem Biophys Res Commun ; 422(4): 573-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22588172

ABSTRACT

We have reported that, in addition to recapitulating the classical human Gaucher disease (GD1) phenotype, deletion of the glucocerebrosidase (GBA1) gene in mice results in the dysfunction of a diverse population of immune cells. Most of immune-related, non-classical features of GD1, including gammopathies and autoimmune diathesis, are resistant to macrophage-directed therapies. This has prompted a search for newer agents for human GD1. Here, we used high-density microarray on splenic and liver cells from affected GBA1(-/-) mice to establish a gene "signature", which was then utilized to interrogate the Broad Institute database, CMAP. Computational connectivity mapping of disease and drug pairs through CMAP revealed several highly enriched, non-null, mimic and anti-mimic hits. Most notably, two compounds with anti-helminthic properties, namely albendazole and oxamniquine, were identified; these are particularly relevant for future testing as the expression of chitinases is enhanced in GD1.


Subject(s)
Computational Biology/methods , Drug Discovery/methods , Gaucher Disease/drug therapy , Gaucher Disease/genetics , Genomics/methods , Glucosylceramidase/deficiency , Molecular Mimicry , Albendazole/pharmacology , Animals , Anthelmintics/pharmacology , Disease Models, Animal , Gene Deletion , Glucosylceramidase/genetics , Liver/drug effects , Liver/metabolism , Mice , Oligonucleotide Array Sequence Analysis/methods , Oxamniquine/pharmacology , Spleen/drug effects , Spleen/metabolism
18.
FASEB J ; 25(1): 182-91, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20826544

ABSTRACT

Both overuse and disuse of joints up-regulate matrix metalloproteinases (MMPs) in articular cartilage and cause tissue degradation; however, moderate (physiological) loading maintains cartilage integrity. Here, we test whether CBP/p300-interacting transactivator with ED-rich tail 2 (CITED2), a mechanosensitive transcriptional coregulator, mediates this chondroprotective effect of moderate mechanical loading. In vivo, hind-limb immobilization of Sprague-Dawley rats up-regulates MMP-1 and causes rapid, histologically detectable articular cartilage degradation. One hour of daily passive joint motion prevents these changes and up-regulates articular cartilage CITED2. In vitro, moderate (2.5 MPa, 1 Hz) intermittent hydrostatic pressure (IHP) treatment suppresses basal MMP-1 expression and up-regulates CITED2 in human chondrocytes, whereas high IHP (10 MPa) down-regulates CITED2 and increases MMP-1. Competitive binding and transcription assays demonstrate that CITED2 suppresses MMP-1 expression by competing with MMP transactivator, Ets-1 for its coactivator p300. Furthermore, CITED2 up-regulation in vitro requires the p38δ isoform, which is specifically phosphorylated by moderate IHP. Together, these studies identify a novel regulatory pathway involving CITED2 and p38δ, which may be critical for the maintenance of articular cartilage integrity under normal physical activity levels.


Subject(s)
Cartilage, Articular/metabolism , Joints/physiology , Matrix Metalloproteinase 1/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line , Chondrocytes/metabolism , Gene Expression , Humans , Hydrostatic Pressure , Immunohistochemistry , Male , Matrix Metalloproteinase 1/genetics , Mutation , Protein Binding , Proto-Oncogene Protein c-ets-1/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Repressor Proteins/metabolism , Restraint, Physical , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
19.
Proc Natl Acad Sci U S A ; 106(17): 7149-54, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19369205

ABSTRACT

We report that oxytocin (OT), a primitive neurohypophyseal hormone, hitherto thought solely to modulate lactation and social bonding, is a direct regulator of bone mass. Deletion of OT or the OT receptor (Oxtr) in male or female mice causes osteoporosis resulting from reduced bone formation. Consistent with low bone formation, OT stimulates the differentiation of osteoblasts to a mineralizing phenotype by causing the up-regulation of BMP-2, which in turn controls Schnurri-2 and 3, Osterix, and ATF-4 expression. In contrast, OT has dual effects on the osteoclast. It stimulates osteoclast formation both directly, by activating NF-kappaB and MAP kinase signaling, and indirectly through the up-regulation of RANK-L. On the other hand, OT inhibits bone resorption by mature osteoclasts by triggering cytosolic Ca(2+) release and NO synthesis. Together, the complementary genetic and pharmacologic approaches reveal OT as a novel anabolic regulator of bone mass, with potential implications for osteoporosis therapy.


Subject(s)
Bone and Bones/metabolism , Oxytocin/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/drug effects , Calcium/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteogenesis/drug effects , Oxytocin/deficiency , Oxytocin/genetics , Oxytocin/pharmacology
20.
Cell Metab ; 34(3): 347-349, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235770

ABSTRACT

In this issue of Cell Metabolism, Lu et al. show that chronic liver disease increases the expression and activity of PP2Ac, a phosphatase that downregulates the excretion of lecithin-cholesterol aceyltransferase (LCAT). LCAT, a liver-derived enzyme, protects bone and prevents bone loss, and its lowered levels in progressive liver injury cause hepatic osteodystrophy (HOD) and worsen liver fibrosis. These discoveries open the possibility that recombinant LCAT may be a treatment for both HOD and liver fibrosis.


Subject(s)
Cholesterol , Phosphatidylcholine-Sterol O-Acyltransferase , Cholesterol/metabolism , Disease Progression , Humans , Liver Cirrhosis , Phosphatidylcholine-Sterol O-Acyltransferase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL