Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Calcif Tissue Int ; 106(2): 208-217, 2020 02.
Article in English | MEDLINE | ID: mdl-31673746

ABSTRACT

Bone is a dynamic tissue that site-specifically adapts to the load that it experiences. In response to increasing load, the cortical bone area is increased, mainly through enhanced periosteal bone formation. This increase in area is associated with an increase in the number of bone-forming osteoblasts; however, the origin of the cells involved remains unclear. Alpha-smooth muscle actin (αSMA) is a marker of early osteoprogenitor cells in the periosteum, and we hypothesized that the new osteoblasts that are activated by loading could originate from αSMA-expressing cells. Therefore, we used an in vivo fate-mapping approach in an established axial loading model to investigate the role of αSMA-expressing cells in the load-induced increase in osteoblasts. Histomorphometric analysis was applied to measure the number of cells of different origin on the periosteal surface in the most load-responsive region of the mouse tibia. A single loading session failed to increase the number of periosteal αSMA-expressing cells and osteoblasts. However, in response to multiple episodes of loading, the caudal, but not the cranial, periosteal surface was lined with an increased number of osteoblasts originating from αSMA-expressing cells 5 days after the initial loading session. The proportion of osteoblasts derived from αSMA-labeled progenitors increased by 70% (p < 0.05), and the proportion of αSMA-labeled cells that had differentiated into osteoblasts was doubled. We conclude that αSMA-expressing osteoprogenitors can differentiate and contribute to the increase in periosteal osteoblasts induced by mechanical loading in a site-specific manner.


Subject(s)
Actins/metabolism , Cell Differentiation , Osteoblasts/physiology , Stem Cells/physiology , Weight-Bearing/physiology , Animals , Cell Proliferation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/physiology , Periosteum/cytology , Stem Cells/metabolism , Stress, Mechanical , Tibia
2.
Osteoporos Int ; 29(9): 2161, 2018 09.
Article in English | MEDLINE | ID: mdl-29987344

ABSTRACT

This article was originally published under a CC BY-NC-ND 4.0 license, but has now been made available under a CC BY 4.0 license. The PDF and HTML versions of the paper have been modified accordingly.

3.
J Endocrinol ; 238(2): 129-136, 2018 08.
Article in English | MEDLINE | ID: mdl-29848607

ABSTRACT

Estrogen treatment has positive effects on the skeleton, and we have shown that estrogen receptor alpha (ERα) expression in cells of hematopoietic origin contributes to a normal estrogen treatment response in bone tissue. T lymphocytes are implicated in the estrogenic regulation of bone mass, but it is not known whether T lymphocytes are direct estrogen target cells. Therefore, the aim of this study was to determine the importance of ERα expression in T lymphocytes for the estrogenic regulation of the skeleton using female mice lacking ERα expression specifically in T lymphocytes (Lck-ERα-/-) and ERαflox/flox littermate (control) mice. Deletion of ERα expression in T lymphocytes did not affect bone mineral density (BMD) in sham-operated Lck-ERα-/- compared to control mice, and ovariectomy (ovx) resulted in a similar decrease in BMD in control and Lck-ERα-/- mice compared to sham-operated mice. Furthermore, estrogen treatment of ovx Lck-ERα-/- led to an increased BMD that was indistinguishable from the increase seen after estrogen treatment of ovx control mice. Detailed analysis of both the appendicular (femur) and axial (vertebrae) skeleton showed that both trabecular and cortical bone parameters responded to a similar extent regardless of the presence of ERα in T lymphocytes. In conclusion, ERα expression in T lymphocytes is dispensable for normal estrogenic regulation of bone mass in female mice.


Subject(s)
Bone and Bones/drug effects , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , T-Lymphocytes/metabolism , Animals , Bone Density/drug effects , Bone Density/genetics , Bone and Bones/metabolism , Estrogen Receptor alpha/metabolism , Female , Gene Expression , Gene Silencing , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Bone ; 112: 10-18, 2018 07.
Article in English | MEDLINE | ID: mdl-29635039

ABSTRACT

Glucocorticoid treatment, a major cause of drug-induced osteoporosis and fractures, is widely used to treat inflammatory conditions and diseases. By contrast, mechanical loading increases bone mass and decreases fracture risk. With these relationships in mind, we investigated whether mechanical loading interacts with GC treatment in bone. Three-month-old female C57BL/6 mice were treated with high-dose prednisolone (15 mg/60 day pellets/mouse) or vehicle for two weeks. During the treatment, right tibiae were subjected to short periods of cyclic compressive loading three times weekly, while left tibiae were used as physiologically loaded controls. The bones were analyzed using peripheral quantitative computed tomography, histomorphometry, real-time PCR, three-point bending and Fourier transform infrared micro-spectroscopy. Loading alone increased trabecular volumetric bone mineral density (vBMD), cortical thickness, cortical area, osteoblast-associated gene expression, osteocyte- and osteoclast number, and bone strength. Prednisolone alone decreased cortical area and thickness and osteoblast-associated gene expression. Importantly, prednisolone treatment decreased the load-induced increase in trabecular vBMD by 57% (p < 0.001) and expression of osteoblast-associated genes, while completely abolishing the load-induced increase in cortical area, cortical thickness, number of osteocytes and osteoclasts, and bone strength. When combined, loading and prednisolone decreased the collagen content. In conclusion, high-dose prednisolone treatment strongly inhibits the loading-induced increase in trabecular BMD, and abolishes the loading-induced increase in cortical bone mass. This phenomenon could be due to prednisolone inhibition of osteoblast differentiation and function.


Subject(s)
Osteogenesis/drug effects , Prednisolone/pharmacology , Anabolic Agents/pharmacology , Animals , Cancellous Bone/drug effects , Cancellous Bone/physiology , Collagen/metabolism , Female , Gene Expression Regulation/drug effects , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/metabolism , Osteogenesis/genetics , Weight-Bearing/physiology
5.
J Mol Endocrinol ; 58(2): 105-111, 2017 02.
Article in English | MEDLINE | ID: mdl-28057769

ABSTRACT

Estradiol (E2) signaling via estrogen receptor alpha (ERα) is important for the male skeleton as demonstrated by ERα inactivation in both mice and man. ERα mediates estrogenic effects not only by translocating to the nucleus and affecting gene transcription but also by extra-nuclear actions e.g., triggering cytoplasmic signaling cascades. ERα contains various domains, and the role of activation function 1 (ERαAF-1) is known to be tissue specific. The aim of this study was to determine the importance of extra-nuclear estrogen effects for the skeleton in males and to determine the role of ERαAF-1 for mediating these effects. Five-month-old male wild-type (WT) and ERαAF-1-inactivated (ERαAF-10) mice were orchidectomized and treated with equimolar doses of 17ß-estradiol (E2) or an estrogen dendrimer conjugate (EDC), which is incapable of entering the nucleus and thereby only initiates extra-nuclear ER actions or their corresponding vehicles for 3.5 weeks. As expected, E2 treatment increased cortical thickness and trabecular bone volume per total volume (BV/TV) in WT males. EDC treatment increased cortical thickness in WT males, whereas no effect was detected in trabecular bone. In ERαAF-10 males, E2 treatment increased cortical thickness, but did not affect trabecular bone. Interestingly, the effect of EDC on cortical bone was abolished in ERαAF-10 mice. In conclusion, extra-nuclear estrogen signaling affects cortical bone mass in males, and this effect is dependent on a functional ERαAF-1. Increased knowledge regarding estrogen signaling mechanisms in the regulation of the male skeleton may aid the development of new treatment options for male osteoporosis.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Protein Domains , Animals , Biomarkers , Bone Remodeling/drug effects , Bone Resorption/blood , Bone Resorption/metabolism , Estrogen Receptor alpha/chemistry , Male , Mice , Osteogenesis/drug effects , Protein Multimerization
6.
Osteoporos Int ; 28(3): 1121-1131, 2017 03.
Article in English | MEDLINE | ID: mdl-27921145

ABSTRACT

Loading increases bone mass and strength in a site-specific manner; however, possible effects of loading on bone matrix composition have not been evaluated. Site-specific structural and material properties of mouse bone were analyzed on the macro- and micro/molecular scale in the presence and absence of axial loading. The response of bone to load is heterogeneous, adapting at molecular, micro-, and macro-levels. INTRODUCTION: Osteoporosis is a degenerative disease resulting in reduced bone mineral density, structure, and strength. The overall aim was to explore the hypothesis that changes in loading environment result in site-specific adaptations at molecular/micro- and macro-scale in mouse bone. METHODS: Right tibiae of adult mice were subjected to well-defined cyclic axial loading for 2 weeks; left tibiae were used as physiologically loaded controls. The bones were analyzed with µCT (structure), reference point indentation (material properties), Raman spectroscopy (chemical), and small-angle X-ray scattering (mineral crystallization and structure). RESULTS: The cranial and caudal sites of tibiae are structurally and biochemically different within control bones. In response to loading, cranial and caudal sites increase in cortical thickness with reduced mineralization (-14 and -3%, p < 0.01, respectively) and crystallinity (-1.4 and -0.3%, p < 0.05, respectively). Along the length of the loaded bones, collagen content becomes more heterogeneous on the caudal site and the mineral/collagen increases distally at both sites. CONCLUSION: Bone structure and composition are heterogeneous, finely tuned, adaptive, and site-specifically responsive at the micro-scale to maintain optimal function. Manipulation of this heterogeneity may affect bone strength, relative to specific applied loads.


Subject(s)
Adaptation, Physiological/physiology , Tibia/physiology , Weight-Bearing/physiology , Animals , Calcification, Physiologic/physiology , Collagen/analysis , Compressive Strength/physiology , Female , Mice, Inbred C57BL , Spectrum Analysis, Raman/methods , Tibia/chemistry , Tibia/diagnostic imaging , X-Ray Microtomography/methods
7.
Endocrinology ; 157(8): 3242-52, 2016 08.
Article in English | MEDLINE | ID: mdl-27254004

ABSTRACT

Estrogens are important regulators of bone mass and their effects are mainly mediated via estrogen receptor (ER)α. Central ERα exerts an inhibitory role on bone mass. ERα is highly expressed in the arcuate (ARC) and the ventromedial (VMN) nuclei in the hypothalamus. To test whether ERα in proopiomelanocortin (POMC) neurons, located in ARC, is involved in the regulation of bone mass, we used mice lacking ERα expression specifically in POMC neurons (POMC-ERα(-/-)). Female POMC-ERα(-/-) and control mice were ovariectomized (OVX) and treated with vehicle or estradiol (0.5 µg/d) for 6 weeks. As expected, estradiol treatment increased the cortical bone thickness in femur, the cortical bone mechanical strength in tibia and the trabecular bone volume fraction in both femur and vertebrae in OVX control mice. Importantly, the estrogenic responses were substantially increased in OVX POMC-ERα(-/-) mice compared with the estrogenic responses in OVX control mice for cortical bone thickness (+126 ± 34%, P < .01) and mechanical strength (+193 ± 38%, P < .01). To test whether ERα in VMN is involved in the regulation of bone mass, ERα was silenced using an adeno-associated viral vector. Silencing of ERα in hypothalamic VMN resulted in unchanged bone mass. In conclusion, mice lacking ERα in POMC neurons display enhanced estrogenic response on cortical bone mass and mechanical strength. We propose that the balance between inhibitory effects of central ERα activity in hypothalamic POMC neurons in ARC and stimulatory peripheral ERα-mediated effects in bone determines cortical bone mass in female mice.


Subject(s)
Bone Density/drug effects , Cortical Bone/drug effects , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Hypothalamus/drug effects , Neurons/drug effects , Pro-Opiomelanocortin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Cortical Bone/metabolism , Female , Hypothalamus/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Pro-Opiomelanocortin/genetics
8.
Tissue Cell ; 47(5): 439-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26337726

ABSTRACT

Disk-degeneration is believed a major cause for lumbar pain. Previously, potential stem cell niches in the intervertebral disk (IVD) region, located adjacent to epiphyseal plate, was reported. The aim of the study was to examine migration of mesenchymal stem cells (MSCs), extracellular matrix (ECM) architecture in a potential cellular migration route (CMR; area located between the niche and IVD) and in the IVD in non-degenerated lapine- and in human degenerated IVD tissues. Human MSCs (n=3), human degenerated IVD tissues (n=10) and lapine IVDs (n=10) were collected. The samples were examined by immunohistochemistry for stem cell markers; CD90, OCT3/4, pre-chondrocytic marker; GDF5, catabolic markers; MMP9, MMP13, inflammatory marker; IL1R, cellular migration markers; SNAI1, SNAI2, adhesion markers; ß1-INTEGRIN and DDR2. In addition, gene-expression analyses (Real time PCR) were performed on additional samples. Further, time lapse studies were performed with hMSCs cultured on aligned COLL-I-fibers-coated glass-slides in DMEM-LG, 10% human serum containing fibroblast growth factor (bFGF). Presence of stem cells (CD90+, OCT3/4+), pre-chondocytic cells (GDF5+) and cells positive for migration markers (SNAI1+, SNAI2+), catabolic markers (MMP9+, MMP13+), inflammatory marker (IL1R+), adhesion markers (DDR2+, B1-INTEGRIN+) were detected (gene- and protein level) in investigated CMR and IVD regions. In the time lapse studies, MSCs alignment and protrusions were observed orientated in the same direction as collagen fibers. Results display influence of ECM collagen architecture and collagen fiber spatial direction on migration of stem cells. The results can be useful when developing tissue-engineering strategies for disk-degeneration.


Subject(s)
Cell Movement/physiology , Extracellular Matrix/metabolism , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc/cytology , Mesenchymal Stem Cells/cytology , Animals , Biomarkers/metabolism , Cells, Cultured , Female , Growth Plate/metabolism , Humans , Intervertebral Disc/metabolism , Low Back Pain/metabolism , Rabbits
9.
Cell Mol Life Sci ; 70(21): 4023-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23516016

ABSTRACT

Estrogens are important endocrine regulators of skeletal growth and maintenance in both females and males. Studies have demonstrated that the estrogen receptor (ER)-α is the main mediator of these estrogenic effects in bone. Therefore, estrogen signaling via ERα is a target both for affecting longitudinal bone growth and bone remodeling. However, treatment with estradiol (E2) leads to an increased risk of side effects such as venous thromboembolism and breast cancer. Thus, an improved understanding of the signaling pathways of ERα will be essential in order to find better bone specific treatments with minimal adverse effects for different estrogen-related bone disorders. This review summarizes the recent data regarding the intracellular signaling mechanisms, in vivo, mediated by the ERα activation functions (AFs), AF-1 and AF-2, and the effect on bone, growth plate and other estrogen responsive tissues. In addition, we review the recent cell-specific ERα-deleted mouse models lacking ERα specifically in neuronal cells or growth plate cartilage. The newly characterized signaling pathways of estrogen, described in this review, provide a better understanding of the ERα signaling pathways, which may facilitate the design of new, bone-specific treatment strategies with minimal adverse effects.


Subject(s)
Bone Development/physiology , Bone and Bones/metabolism , Cartilage/metabolism , Estrogen Receptor alpha/physiology , Growth Plate/metabolism , Animals , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Humans , Male , Mice , Neurons/metabolism , Osteoclasts/metabolism , Signal Transduction
10.
Am J Physiol Endocrinol Metab ; 302(11): E1381-9, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22414805

ABSTRACT

High estradiol levels in late puberty induce growth plate closure and thereby cessation of growth in humans. In mice, the growth plates do not fuse after sexual maturation, but old mice display reduced longitudinal bone growth and high-dose estradiol treatment induces growth plate closure. Estrogen receptor (ER)-α stimulates gene transcription via two activation functions (AFs), AF-1 and AF-2. To evaluate the role of ERα and its AF-1 for age-dependent reduction in longitudinal bone growth and growth plate closure, female mice with inactivation of ERα (ERα(-/-)) or ERαAF-1 (ERαAF-1(0)) were evaluated. Old (16- to 19-mo-old) female ERα(-/-) mice showed continued substantial longitudinal bone growth, resulting in longer bones (tibia: +8.3%, P < 0.01) associated with increased growth plate height (+18%, P < 0.05) compared with wild-type (WT) mice. In contrast, the longitudinal bone growth ceased in old ERαAF-1(0) mice (tibia: -4.9%, P < 0.01). Importantly, the proximal tibial growth plates were closed in all old ERαAF-1(0) mice while they were open in all WT mice. Growth plate closure was associated with a significantly altered balance between chondrocyte proliferation and apoptosis in the growth plate. In conclusion, old female ERα(-/-) mice display a prolonged and enhanced longitudinal bone growth associated with increased growth plate height, resembling the growth phenotype of patients with inactivating mutations in ERα or aromatase. In contrast, ERαAF-1 deletion results in a hyperactive ERα, altering the chondrocyte proliferation/apoptosis balance, leading to growth plate closure. This suggests that growth plate closure is induced by functions of ERα that do not require AF-1 and that ERαAF-1 opposes growth plate closure.


Subject(s)
Estrogen Receptor alpha/physiology , Growth Plate/physiology , Trans-Activators/physiology , Absorptiometry, Photon , Aging/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Bone Development/drug effects , Cell Proliferation , Chondrocytes/physiology , DNA Primers , Estradiol/blood , Estrogen Receptor alpha/genetics , Female , Growth Plate/anatomy & histology , Immunohistochemistry , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proliferating Cell Nuclear Antigen/metabolism , Sexual Maturation/physiology , Tibia/growth & development , Trans-Activators/genetics
11.
Proc Natl Acad Sci U S A ; 108(15): 6288-93, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21444817

ABSTRACT

The bone-sparing effect of estrogen is primarily mediated via estrogen receptor-α (ERα), which stimulates target gene transcription through two activation functions (AFs), AF-1 in the N-terminal and AF-2 in the ligand binding domain. To evaluate the role of ERα AF-1 and ERα AF-2 for the effects of estrogen in bone in vivo, we analyzed mouse models lacking the entire ERα protein (ERα(-/-)), ERα AF-1 (ERαAF-1(0)), or ERα AF-2 (ERαAF-2(0)). Estradiol (E2) treatment increased the amount of both trabecular and cortical bone in ovariectomized (OVX) WT mice. Neither the trabecular nor the cortical bone responded to E2 treatment in OVX ERα(-/-) or OVX ERαAF-2(0) mice. OVX ERαAF-1(0) mice displayed a normal E2 response in cortical bone but no E2 response in trabecular bone. Although E2 treatment increased the uterine and liver weights and reduced the thymus weight in OVX WT mice, no effect was seen on these parameters in OVX ERα(-/-) or OVX ERαAF-2(0) mice. The effect of E2 in OVX ERαAF-1(0) mice was tissue-dependent, with no or weak E2 response on thymus and uterine weights but a normal response on liver weight. In conclusion, ERα AF-2 is required for the estrogenic effects on all parameters evaluated, whereas the role of ERα AF-1 is tissue-specific, with a crucial role in trabecular bone and uterus but not cortical bone. Selective ER modulators stimulating ERα with minimal activation of ERα AF-1 could retain beneficial actions in cortical bone, constituting 80% of the skeleton, while minimizing effects on reproductive organs.


Subject(s)
Bone and Bones/physiology , Estrogen Receptor alpha/physiology , Estrogens/physiology , Animals , Bone Density , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , Mice , Mice, Mutant Strains , Organ Size , Radiography , Selective Estrogen Receptor Modulators/pharmacology , Thymus Gland/anatomy & histology , Thymus Gland/drug effects , Thymus Gland/physiology , Transcriptional Activation , Uterus/anatomy & histology , Uterus/drug effects , Uterus/physiology
12.
Am J Physiol Endocrinol Metab ; 296(3): E490-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19088255

ABSTRACT

In vitro studies suggest that the membrane G protein-coupled receptor GPR30 is a functional estrogen receptor (ER). The aim of the present study was to determine the possible in vivo role of GPR30 as a functional ER primarily for the regulation of skeletal parameters, including bone mass and longitudinal bone growth, but also for some other well-known estrogen-regulated parameters, including uterine weight, thymus weight, and fat mass. Three-month-old ovariectomized (OVX) GPR30-deficient mice (GPR30(-/-)) and wild-type (WT) mice were treated with either vehicle or increasing doses of estradiol (E(2); 0, 30, 70, 160, or 830 ng.mouse(-1).day(-1)). Body composition [bone mineral density (BMD), fat mass, and lean mass] was analyzed by dual-energy-X ray absorptiometry, while the cortical and trabecular bone compartments were analyzed by peripheral quantitative computerized tomography. Quantitative histological analyses were performed in the distal femur growth plate. Bone marrow cellularity and distribution were analyzed using a fluorescence-activated cell sorter. The estrogenic responses on most of the investigated parameters, including increase in bone mass (total body BMD, spine BMD, trabecular BMD, and cortical bone thickness), increase in uterine weight, thymic atrophy, fat mass reduction, and increase in bone marrow cellularity, were similar for all of the investigated E(2) doses in WT and GPR30(-/-) mice. On the other hand, E(2) treatment reduced longitudinal bone growth, reflected by decreased femur length and distal femur growth plate height, in the WT mice but not in the GPR30(-/-) mice compared with vehicle-treated mice. These in vivo findings demonstrate that GPR30 is not required for normal estrogenic responses on several major well-known estrogen-regulated parameters. In contrast, GPR30 is required for a normal estrogenic response in the growth plate.


Subject(s)
Bone Development/physiology , Estrogens/metabolism , Ovariectomy , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Adipose Tissue/anatomy & histology , Adipose Tissue/growth & development , Animals , Bone Density , Female , Femur/cytology , Femur/growth & development , Growth Plate/cytology , Growth Plate/growth & development , Mice , Mice, Mutant Strains , Organ Size , Receptors, Estrogen/metabolism , Thymus Gland/anatomy & histology , Thymus Gland/growth & development , Uterus/anatomy & histology , Uterus/growth & development
13.
Endocrinology ; 148(12): 5688-95, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761761

ABSTRACT

Estrogen has bone protective effects, but the exact mechanism behind these effects remains unclear. The aim of the present study was to identify the primary target cells in bone for the classical genomic effects of estrogens in vivo. For this purpose we have used reporter mice with a luciferase gene under the control of three estrogen-responsive elements (EREs), enabling detection of in vivo activation of gene transcription. Three-month-old ovariectomized mice were treated with a single dose (50 mug/kg) 17beta-estradiol (E2). Luciferase activity was analyzed in several tissues and in different bone marrow-derived lymphocyte enriched/depleted preparations using MacsMouse CD19 (for B lymphocytes) or CD90 (for T lymphocytes) MicroBeads (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany). Histological characterization of cells with high luciferase content was performed using immunohistochemistry. Both cortical bone and bone marrow displayed a rapid (within 1 h) and pronounced E2-induced increase in luciferase activity. The luciferase activity in total bone marrow and in bone marrow depleted of lymphocytes was increased six to eight times more than in either B-lymphocyte or T-lymphocyte enriched cell fractions 4 h after the E2 injection, demonstrating that mature lymphocytes are not major direct targets for the genomic effect of estrogens in bone. Immunohistochemistry identified clear luciferase staining in hypertrophic growth plate chondrocytes, megakaryocytes, osteoblasts, and lining cells, whereas no staining was seen in proliferative chondrocyte. Although most of the osteocytes did not display any detectable luciferase staining, a subpopulation of osteocytes both in cortical and trabecular bone stained positive for luciferase. In conclusion, hypertrophic growth plate chondrocytes, megakaryocytes, osteoblasts, lining cells, and a subpopulation of osteocytes were identified to respond to estrogen via the classical ERE-mediated genomic pathway in bone. Furthermore, our findings indicate that possible direct estrogenic effects on the majority of osteocytes, not staining positive for luciferase, on proliferative chondrocytes and on mature lymphocytes are mediated by non-ERE actions.


Subject(s)
Bone and Bones/drug effects , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone and Bones/cytology , Bone and Bones/metabolism , Estradiol/administration & dosage , Estradiol/pharmacology , Estrogens/administration & dosage , Female , Flow Cytometry , Growth Plate/cytology , Growth Plate/drug effects , Growth Plate/metabolism , Immunohistochemistry , Luciferases/genetics , Luciferases/metabolism , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Megakaryocytes/cytology , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Ovariectomy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics
14.
J Clin Endocrinol Metab ; 91(12): 5029-37, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16926255

ABSTRACT

CONTEXT: SHBG regulates free sex steroid levels, which in turn regulate skeletal homeostasis. Twin studies have demonstrated that genetic factors largely account for interindividual variation in SHBG levels. Glucuronidated androgen metabolites have been proposed as markers of androgenic activity. OBJECTIVE: Our objective was to investigate whether polymorphisms in the SHBG gene promoter [(TAAAA)(n) microsatellite and rs1799941 single-nucleotide polymorphism] are associated with serum levels of SHBG, sex steroids, or bone mineral density (BMD) in men. DESIGN AND STUDY SUBJECTS: We conducted a population-based study of two cohorts of Swedish men: elderly men (MrOS Sweden; n congruent with 3000; average age, 75.4 yr) and young adult men (GOOD study; n = 1068; average age, 18.9 yr). MAIN OUTCOME MEASURES: We measured serum levels of SHBG, testosterone, estradiol, dihydrotestosterone, 5alpha-androstane-3alpha,17beta-diol glucuronides, androsterone glucuronide, and BMD determined by dual-energy x-ray absorptiometry. RESULTS: In both cohorts, (TAAAA)(n) and rs1799941 genotypes were associated with serum levels of SHBG (P < 0.001), dihydrotestosterone (P < 0.05), and 5alpha-androstane-3alpha,17beta-diol glucuronides (P < 0.05). In the elderly men, they were also associated with testosterone and BMD at all hip bone sites. The genotype associated with high levels of SHBG was also associated with high BMD. Interestingly, male mice overexpressing human SHBG had increased cortical bone mineral content in the femur, suggesting that elevated SHBG levels may cause increased bone mass. CONCLUSIONS: Our findings demonstrate that polymorphisms in the SHBG promoter predict serum levels of SHBG, androgens, and glucuronidated androgen metabolites, and hip BMD in men.


Subject(s)
Androgens/blood , Bone Density/physiology , Polymorphism, Genetic , Sex Hormone-Binding Globulin/analysis , Sex Hormone-Binding Globulin/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Androgens/metabolism , Animals , Genotype , Hip/physiology , Humans , Male , Mice , Mice, Transgenic , Microsatellite Repeats/physiology , Promoter Regions, Genetic
15.
J Endocrinol ; 174(2): 167-78, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12176656

ABSTRACT

Estrogen exerts a variety of important physiological effects, which have been suggested to be mediated via the two known estrogen receptors (ERs), alpha and beta. Three-month-old ovariectomized mice, lacking one or both of the two estrogen receptors, were given estrogen subcutaneously (2.3 micro g/mouse per day) and the effects on different estrogen-responsive parameters, including skeletal effects, were studied. We found that estrogen increased the cortical bone dimensions in both wild-type (WT) and double ER knockout (DERKO) mice. DNA microarray analysis was performed to characterize this effect on cortical bone and it identified four genes that were regulated by estrogen in both WT and DERKO mice. The effect of estrogen on cortical bone in DERKO mice might either be due to remaining ERalpha activity or represent an ERalpha/ERbeta-independent effect. Other effects of estrogen, such as increased trabecular bone mineral density, thymic atrophy, fat reduction and increased uterine weight, were mainly ERalpha mediated.


Subject(s)
Bone and Bones/drug effects , Estrogens/pharmacology , Receptors, Estrogen/physiology , Animals , Bone Density/drug effects , Collagen Type VIII/genetics , Complement C3/genetics , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Integrin-Binding Sialoprotein , Interleukin-3/genetics , Liver/drug effects , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Organ Size/drug effects , Ovariectomy , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Sialoglycoproteins/genetics , Thymus Gland/cytology , Uterus/cytology , Uterus/metabolism , alpha-Macroglobulins/genetics
16.
J Endocrinol ; 171(3): 425-33, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739008

ABSTRACT

Estrogens are important for the male skeleton. Osteoprotegerin (OPG), receptor activator of NF-kappa B ligand (RANKL), interleukin-6 (IL-6), IL-1 and tumor necrosis factor alpha (TNFalpha) have been suggested to be involved in the skeletal effects of estrogen. We treated orchidectomized mice with estradiol for 2 weeks and observed a 143% increase in the trabecular bone mineral density of the distal metaphysis of femur that was associated with a decreased OPG/RANKL mRNA ratio in vertebral bone. A similar decreased OPG/RANKL ratio was also seen after estrogen treatment of ovariectomized female mice. The effect of estrogen receptor (ER) inactivation on the OPG/RANKL ratio was dissected by using intact male mice lacking ER alpha (ERKO), ER beta (BERKO) or both receptors (DERKO). The expression of OPG was increased in ERKO and DERKO but not in BERKO male mice, resulting in an increased OPG/RANKL ratio. Furthermore, serum levels of IL-6 and tartrate-resistant acid phosphatase 5b (TRAP 5b) were decreased in ERKO and DERKO, but not in BERKO male mice. These results demonstrate that ER alpha, but not ER beta, is involved in the regulation of the vertebral OPG/RANKL ratio, serum levels of IL-6 and TRAP 5b in male mice.


Subject(s)
Carrier Proteins , Glycoproteins/metabolism , Interleukin-6/blood , Membrane Glycoproteins , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/physiology , Receptors, Tumor Necrosis Factor/metabolism , Acid Phosphatase/blood , Animals , Biomarkers/blood , Bone Density/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Isoenzymes/blood , Ligands , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Orchiectomy , Osteoprotegerin , Ovariectomy , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Tartrate-Resistant Acid Phosphatase
17.
J Bone Miner Res ; 16(8): 1388-98, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11499861

ABSTRACT

Recently, it has been shown that inactivation of estrogen receptor beta (ER-beta) by gene targeting results in increased cortical bone formation in adolescent female mice. To study the possible involvement of ER-beta in the regulation of the mature skeleton, we have extended the analyses to include 1-year-old ER-beta knockout mice (ER-beta-/-). Male ER-beta-/- mice did not express any significant bone phenotypic alterations at this developmental stage. However, the increase in cortical bone parameters seen already in the adolescent female ER-beta-/- mice was maintained in the older females. The aged female ER-beta-/- mice further exhibited a significantly higher trabecular bone mineral density (BMD) as well as increased bone volume/total volume (BV/TV) compared with wild-type (wt) mice. This was caused by a less pronounced loss of trabecular bone during adulthood in female ER-beta-/- mice. The growth plate width was unaltered in the female ER-beta-/- mice. Judged by the expression of the osteoclast marker tartrate-resistant acid phosphatase (TRAP) and cathepsin K (cat K; reverse-transcription-polymerase chain reaction [RT-PCR]) as well as the serum levels of C-terminal type I collagen cross-linked peptide, bone resorption appeared unaffected. However, an increase in the messenger RNA (mRNA) expression levels of the osteoblast marker core-binding factor alpha1 (Cbfa1) suggested an anabolic effect in bones of old female ER-beta-/- mice. In addition, the mRNA expression of ER-alpha was augmented, indicating a role for ER-alpha in the development of this phenotype. Taken together, the results show that ER-beta is involved in the regulation of trabecular bone during adulthood in female mice and suggest that ER-beta acts in a repressive manner, possibly by counteracting the stimulatory action of ER-alpha on bone formation.


Subject(s)
Osteoporosis/metabolism , Receptors, Estrogen/physiology , Animals , Body Weight , Bone Density , Collagen/blood , Collagen Type I , Cross-Linking Reagents , Disease Models, Animal , Estradiol/blood , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Femur/metabolism , Femur/pathology , Gene Expression , Insulin-Like Growth Factor I/analysis , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteocalcin/blood , Osteoclasts/metabolism , Osteoporosis/pathology , Osteoporosis/prevention & control , Peptides/blood , RNA, Messenger/metabolism , Receptors, Estrogen/genetics
18.
Bone ; 26(2): 117-21, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10678405

ABSTRACT

Estrogens affect bone metabolism, and ovariectomy of rats results in marked bone loss caused by stimulation of osteoclastic bone resorption. Estrogen receptors (ER) have been demonstrated in osteoblasts and bone marrow stromal cells, but their presence in osteoclasts is controversial. Until recently, only one type of ER (now renamed ERalpha) had been identified. After the discovery of a novel ER subtype (ERbeta), it became necessary to re-investigate the ER expression in human and rodent bone. In the present study we examined the expression of ER mRNA in neonatal rat bone. Expression of ER alpha and beta mRNA (RT-PCR) was evident in femurs of 3-week-old male and female rats. In situ hybridization histochemistry of femural bones with digoxigenin labelled riboprobes, as well as radioactively labeled riboprobes, revealed that ERbeta mRNA was predominantly expressed in osteoblasts covering the metaphyseal bone trabecular surface. The presence of ERbeta mRNA in osteoblasts of rat bone suggests that ERbeta is involved in the mechanism of action of estrogens in bone.


Subject(s)
Bone and Bones/metabolism , Receptors, Estrogen/genetics , Animals , Animals, Newborn , Bone and Bones/cytology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Gene Expression , Humans , In Situ Hybridization , Male , Osteoblasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
19.
J Clin Invest ; 104(7): 895-901, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10510330

ABSTRACT

Ovariectomy in young, growing rodents results in decreased trabecular bone mineral density (BMD) and increased radial growth of the cortical bone. Both of these effects are reversed by treatment with estrogen. The aim of the present study was to determine the physiological role of estrogen receptor-beta (ERbeta) on bone structure and bone mineral content (BMC). The BMC was increased in adult (11 weeks old), but not prepubertal (4 weeks old), female ERbeta(-/-) mice compared with wild-type (WT) mice. This increase in BMC in females was not due to increased trabecular BMD, but to an increased cross-sectional cortical bone area associated with a radial bone growth. Male ERbeta(-/-) mice displayed no bone abnormalities compared with WT mice. Ovariectomy decreased the trabecular BMD to the same extent in adult female ERbeta(-/-) mice as in WT mice. The expression levels of osteoblast-associated genes - alpha1(I) collagen, alkaline phosphatase, and osteocalcin mRNAs - were elevated in bone from adult ERbeta(-/-) females compared with WT mice. These observations provide a possible explanation for the increased radial bone growth seen in female mutants, suggesting a repressive function for ERbeta in the regulation of bone growth during female adolescence. In summary, ERbeta is essential for the pubertal feminization of the cortical bone in female mice but is not required for the protective effect of estrogens on trabecular BMD.


Subject(s)
Bone Density/physiology , Bone Development , Receptors, Estrogen/physiology , Aging , Alkaline Phosphatase/genetics , Animals , Body Weight , CD40 Ligand , Collagen/genetics , Estrogen Receptor beta , Female , Femur/growth & development , Gene Expression Regulation, Developmental , Growth Plate/growth & development , Male , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Ovariectomy , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sex Characteristics , Sexual Maturation , Tibia/growth & development
20.
J Steroid Biochem Mol Biol ; 71(3-4): 93-102, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10659697

ABSTRACT

The glucocorticoid receptor (GR) regulates target gene expression in response to corticosteroid hormones. We have investigated the mechanism of transcriptional activation by the GR by studying the role of the receptor interacting protein RIP140. Both in vivo and in vitro protein-protein interaction assays revealed a ligand-dependent interaction between the GR and RIP140. The ligand binding domain of the GR was sufficient for this interaction, while both the N- and C-terminal regions of RIP140 bound to the receptor. In a yeast transactivation assay RIP140 and SRC-1, a member of the steroid receptor coactivator family of proteins, both enhanced the transactivation activity of a GR protein (GRA-1) in which the potent N-terminal tau1 transactivation domain has been deleted. In contrast, in COS-7 cells increasing amounts of RIP140 significantly inhibited GRdeltatau1 function. In cotransfection studies in COS-7 cells, RIP140 also inhibited receptor activity in presence of both SRC-1 and the coactivator protein CBP together. Thus, in yeast cells a stimulation of receptor activity was observed, while in mammalian cells RIP140 repressed GR function. Taken together, these data suggest that, (1) RIP140 is a target protein for the GR and (2) RIP140 can modulate the transactivation activity of the receptor.


Subject(s)
Nuclear Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , COS Cells , CREB-Binding Protein , Histone Acetyltransferases , Humans , In Vitro Techniques , Ligands , Nuclear Proteins/genetics , Nuclear Receptor Coactivator 1 , Nuclear Receptor Interacting Protein 1 , Protein Binding , Receptors, Glucocorticoid/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...