Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(12): 2046-57, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25909889

ABSTRACT

Regulators of G protein signaling (Rgs) have pivotal roles in controlling various cellular processes, such as cell differentiation. How Rgs proteins regulate osteoclast (OC) differentiation, function and bone homeostasis is poorly understood. It was previously demonstrated that Rgs12, the largest protein in the Rgs family, is predominantly expressed in OCs and regulates OC differentiation in vitro. To further understand the role and mechanism of Rgs12 in OC differentiation and bone diseases in vivo, we created OC-targeted Rgs12 knockout mice by using inducible Mx1-Cre and CD11b-Cre. Deletion of Rgs12 in hematopoietic cells or specifically in OC precursors resulted in increased bone mass with decreased OC numbers. Loss of Rgs12 impaired OC differentiation and function with impaired Ca(2+) oscillations and reduced nuclear factor of activated T cells (NFAT) 2 expression. The introduction of wild-type osteoblasts did not rescue the defective osteoclastogenesis. Ectopic expression of NFAT2 rescued defective OC differentiation in CD11b;Rgs12(fl/fl) cells and promoted normal OC differentiation. Moreover, deletion of Rgs12 significantly inhibited pathological osteoclastogenesis and bone destruction in Rgs12-deficient mice that were subjected to ovariectomy and lipodysaccharide for bone loss. Thus our findings demonstrate that Rgs12 is an important regulator in OC differentiation and function and identify Rgs12 as a potential therapeutic target for osteoporosis and inflammation-induced bone loss.


Subject(s)
Bone Remodeling , Bone and Bones/metabolism , RGS Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , CD11b Antigen/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Femur/diagnostic imaging , Femur/metabolism , Femur/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis/drug effects , RGS Proteins/deficiency , RGS Proteins/genetics , X-Ray Microtomography
2.
Biochem Biophys Res Commun ; 392(1): 72-6, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20059964

ABSTRACT

TGFbeta inducible early gene-1 (TIEG) is a member of the Krüppel-like family of transcription factors (KLF10) that plays an important role in TGFbeta mediated Smad signaling. In order to better understand the role of TIEG in bone, we generated TIEG knockout (KO) mice. Calvarial osteoblasts (OBs) isolated from these mice exhibit a reduced ability to support osteoclastogenesis in vitro. Gene expression studies revealed decreased receptor activator of NF-kappaB ligand (RANKL) and increased osteoprotegerin (OPG) expression in TIEG KO OBs, suggesting a potential role for TIEG in regulating the expression of these genes. Since OPG and RANKL are two important regulators of osteoclast (OC) differentiation, we sought to determine if TIEG directly regulates their expression. Luciferase constructs, containing fragments of either the mouse OPG promoter (-1486 to +133 bp) or the RANKL promoter (-2000 to +1 bp) were each cloned into the pGL3 basic reporter vector and transiently transfected into TIEG KO calvarial OBs with and without a TIEG expression vector. No significant changes in the activity of the RANKL promoter were detected in the presence of TIEG. However, OPG promoter activity was inhibited in the presence of TIEG protein suggesting that TIEG directly represses the expression of OPG in OBs. In order to determine the region of this promoter through which TIEG acts, sequential 5'-deletion constructs were generated. Transient transfection of these constructs revealed that the TIEG regulatory element(s) reside within a 200 bp region of the OPG promoter. Transient ChIP analyses, using a TIEG-specific antibody, revealed that TIEG binds to this region of the OPG promoter. Since we have previously shown that TIEG regulates target gene expression through Sp-1 sites, we examined this region of the OPG promoter for potential TIEG binding elements and identified four potential Sp-1 binding sites. Site-directed mutagenesis was used to determine if TIEG utilizes these Sp-1 elements to regulate the activity of the OPG promoter. The data demonstrate that two Sp-1 sites are likely to be involved in TIEG's repression of the OPG promoter. Taken together, these results confirm that TIEG directly binds to and inhibits OPG promoter activity in OBs, partially explaining the inability of TIEG KO OBs to fully support osteoclast differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Osteoblasts/metabolism , Osteoprotegerin/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Differentiation/genetics , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , RANK Ligand/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics
3.
Osteoporos Int ; 20(5): 761-9, 2009 May.
Article in English | MEDLINE | ID: mdl-18769961

ABSTRACT

UNLABELLED: In bone marrow aspirates from postmenopausal women, systemic estrogen treatment decreased differentiation of mononuclear progenitor cells toward a more mature osteoclast phenotype. This was not associated with changes in surface receptor for proresorptive cytokines. INTRODUCTION: Although mechanisms by which estrogen (E) decreases bone resorption have been extensively studied in rodents, little information is available in humans. METHODS: In bone marrow aspirates from 34 early postmenopausal women randomly assigned to receive 4 weeks of treatment (100 microg/day of transdermal 17beta-estradiol) or no treatment, we assessed osteoclast differentiation and surface receptors using flow cytometry with fluorescent-labeled specific antibodies. RESULTS: E treatment decreased (P < 0.05) the proportion of bone marrow mononuclear cells (BMMNCs) expressing the calcitonin receptor (CTR), a late osteoclast phenotype marker. There was an increase in c-Fms concentration in osteoclast lineage cells (P < 0.05) and in the proportion of BMMNCs expressing TNFR2 (P < 0.05), but there were no significant effects on other surface receptors for proresorptive factors (RANK, TNFR1, TREM2, or OSCAR). Changes in serum CTx and TRAP 5b, markers for bone resorption, correlated directly (P < 0.05) with the proportion of BMMNCs expressing CTR and, for TRAP 5b only, TNFR2 and inversely with c-Fms concentration (all P < 0.05). CONCLUSION: E reduces bone resorption, in part, by decreasing differentiation of BMMNCs into mature osteoclasts. This action cannot be explained by decreased concentrations of surface receptors for proresorptive factors. The roles of increases in c-Fms concentration and the proportion of TNFR2((+)) cells are unclear.


Subject(s)
Bone Marrow Cells/drug effects , Bone Resorption/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Osteoclasts/drug effects , Postmenopause , Adult , Aged , Bone Marrow Cells/metabolism , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cytokines/metabolism , Female , Flow Cytometry , Humans , Middle Aged , Osteoclasts/cytology , Osteoclasts/metabolism , Postmenopause/metabolism , Receptors, Calcitonin/metabolism
4.
Bone ; 42(6): 1025-31, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18396127

ABSTRACT

TGFbeta inducible early gene-1 (TIEG) was originally cloned from human osteoblasts (OB) and has been shown to play an important role in TGFbeta/Smad signaling, regulation of gene expression and OB growth and differentiation. To better understand the biological role of TIEG in the skeleton, we have generated congenic TIEG-null (TIEG(-/-)) mice in a pure C57BL/6 background. Through the use of DXA and pQCT analysis, we have demonstrated that the femurs and tibias of two-month-old female TIEG(-/-) mice display significant decreases in total bone mineral content, density, and area relative to wild-type (WT) littermates. However, no differences were observed for any of these bone parameters in male mice. Further characterization of the bone phenotype of female TIEG(-/-) mice involved mechanical 3-point bending tests, micro-CT, and histomorphometric analyses of bone. The 3-point bending tests revealed that the femurs of female TIEG(-/-) mice have reduced strength with increased flexibility compared to WT littermates. Micro-CT analysis of femurs of two-month-old female TIEG(-/-) mice revealed significant decreases in cortical bone parameters compared to WT littermates. Histomorphometric evaluation of the distal femur revealed that female TIEG(-/-) mice also display a 31% decrease in cancellous bone area, which is primarily due to a decrease in trabecular number. At the cellular level, female TIEG(-/-) mice exhibit a 42% reduction in bone formation rate which is almost entirely due to a reduction in double labeled perimeter. Differences in mineral apposition rate were not detected between WT and TIEG(-/-) mice. Taken together, these findings suggest that female TIEG(-/-) mice are osteopenic mainly due to a decrease in the total number of functional/mature OBs.


Subject(s)
Bone Diseases, Metabolic/physiopathology , DNA-Binding Proteins/metabolism , Femur , Tibia , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Femur/cytology , Femur/pathology , Femur/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/physiology , Phenotype , Sex Factors , Signal Transduction/physiology , Stress, Mechanical , Tibia/cytology , Tibia/pathology , Tibia/physiology , Transcription Factors/genetics
5.
Osteoporos Int ; 19(9): 1323-30, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18274695

ABSTRACT

UNLABELLED: One-year treatment of osteoporotic postmenopausal women with transdermal estrogen resulted in significant decreases in bone marrow adipocyte volume and prevented increases in adipocyte number as compared to placebo-treated controls. Estrogen treatment also prevented increases in mean adipocyte size over 1 year. INTRODUCTION: Aging is associated not only with bone loss but also with increases in bone marrow adipocytes. Since osteoblasts and adipocytes are derived from a common precursor, it is possible that with aging, there is a preferential "switch" in commitment of this precursor to the adipocyte over the osteoblast lineage. We tested the hypothesis that the apparent "age-related" increase in marrow adipocytes is due, at least in part, to estrogen (E) deficiency. METHODS: Reanalysis of bone biopsies from a randomized, placebo-controlled trial involving 56 postmenopausal osteoporotic women (mean age, 64 years) treated either with placebo (PL, n = 27) or transdermal estradiol (0.1 mg/d, n = 29) for 1 year. RESULTS: Adipocyte volume/tissue volume (AV/TV) and adipocyte number (Ad#) increased (by 20%, P < 0.05) in the PL group, but were unchanged (Ad#) or decreased (AV/TV, by -24%, P < 0.001) in the E group. E treatment also prevented increases in mean adipocyte size over 1 year. CONCLUSIONS: These findings represent the first in vivo demonstration in humans that not only ongoing bone loss, but also the increase in bone marrow adipocyte number and size in postmenopausal osteoporotic women may be due, at least in part, to E deficiency.


Subject(s)
Adipocytes/drug effects , Bone Marrow Cells/drug effects , Estradiol/pharmacology , Estrogen Replacement Therapy/methods , Osteoporosis, Postmenopausal/pathology , Adipocytes/pathology , Administration, Cutaneous , Aged , Anthropometry , Biopsy , Bone Density/drug effects , Bone Marrow Cells/pathology , Cell Count , Cell Size/drug effects , Estradiol/blood , Female , Humans , Leptin/blood , Lipids/blood , Middle Aged , Osteoporosis, Postmenopausal/blood
6.
J Cell Biochem ; 103(2): 383-92, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17541956

ABSTRACT

It is well established that E(2) and TGFbeta have major biological effects in multiple tissues, including bone. The signaling pathways through which these two factors elicit their effects are well documented. However, the interaction between these two pathways and the potential consequences of cross-talk between E(2) and TGFbeta continue to be elucidated. In this prospectus, we present known and potential roles of TIEG, Runx2, and other transcription factors as important mediators of signaling between these two pathways.


Subject(s)
Bone and Bones/metabolism , Core Binding Factor Alpha 1 Subunit/physiology , DNA-Binding Proteins/physiology , Early Growth Response Transcription Factors/physiology , Estrogens/physiology , Kruppel-Like Transcription Factors/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Animals , Bone Remodeling/physiology , Dimerization , Female , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Protein Isoforms/genetics , Protein Isoforms/physiology , Rats , Transcription, Genetic , Transforming Growth Factor beta/deficiency , Transforming Growth Factor beta/genetics
7.
J Cell Biochem ; 99(2): 425-34, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16619269

ABSTRACT

2-Methoxyestradiol (2-ME), a naturally occurring metabolite of 17beta-estradiol, is highly cytotoxic to a wide range of tumor cells but is harmless to most normal cells. However, 2-ME prevented bone loss in ovariectomized rats, suggesting it inhibits bone resorption. These studies were performed to determine the direct effects of 2-ME on cultured osteoclasts. 2-ME (2 microM) reduced osteoclast number by more than 95% and induced apoptosis in three cultured osteoclast model systems (RAW 264.7 cells cultured with RANKL, marrow cells co-cultured with stromal support cells, and spleen cells cultured without support cells in media supplemented with RANKL and macrophage colony stimulating factor (M-CSF)). The 2-ME-mediated effect was ligand specific; 2-hydroxyestradiol (2-OHE), the immediate precursor to 2-ME, exhibited less cytotoxicity; and 2-methoxyestrone (2-MEOE1) the estrone analog of 2-ME, was not cytotoxic. Co-treatment with ICI 182,780 did not antagonize 2-ME, suggesting that the cytotoxicity was not estrogen receptor-dependent. 2-ME-induced cell death in RAW 264.7 cells coincided with an increase in gene expression of cytokines implicated in inhibition of differentiation and induction of apoptosis. In addition, the 2-ME-mediated decrease in cell survival was partially inhibited by anti-lymphotoxin(LT)beta antibodies, suggesting that 2-ME-dependent effects involve LTbeta. These results suggest that 2-ME could be useful for treating skeletal diseases in which bone resorption is increased, such as postmenopausal osteoporosis and cancer metastasis to bone.


Subject(s)
Estradiol/analogs & derivatives , Osteoclasts/cytology , Osteoclasts/drug effects , 2-Methoxyestradiol , Animals , Apoptosis/drug effects , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Female , Fulvestrant , Lymphotoxin-alpha/antagonists & inhibitors , Lymphotoxin-beta , Membrane Proteins/antagonists & inhibitors , Mice , Osteoclasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
8.
J Musculoskelet Neuronal Interact ; 3(4): 363-6; discussion 381, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15758324
9.
J Bone Miner Res ; 16(6): 1028-36, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11393779

ABSTRACT

To investigate the role of the pp60src signaling pathway in osteoclast activity, we have used dominant negative pp60src, c-ras, and c-raf expression vectors to individually disrupt their functions in osteoclasts. Osteoclasts were transiently transfected and secretions of cathepsin B/K and tartrate-resistant acid phosphatase (TRAP) were monitored. Expression of these constructs increased secretion of lysosomal enzymes. In contrast, constitutively active pp60src expression caused decreased lysosomal enzyme secretion. Osteoclasts respond to 17-beta estradiol (17betaE2) treatment with decreased lysosomal enzyme secretion. Therefore, we investigated the effects of E2 on pp60src kinase activity and observed an E2 time- and dose-dependent decrease in cytoskeletal membrane-associated pp60src tyrosine kinase activity. We have shown that estrogen decreases lysosomal enzyme gene expression and secretion; so we have examined the effects of the expression constructs on estrogen regulation of enzyme secretion. Constitutively active pp60src blocked E2 effects on secretion whereas expression of dominant negative pp60src, c-Ras, or c-Raf enhanced E2 effects. These data support that the kinase domain of cytoskeletal-associated pp60src is likely to be involved in the regulation of lysosomal enzyme secretion.


Subject(s)
Estradiol/analogs & derivatives , Estrogens/metabolism , Lysosomes/enzymology , Osteoclasts/metabolism , Signal Transduction , src-Family Kinases/metabolism , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Cells, Cultured , Chickens , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens/pharmacology , Fulvestrant , Genes, ras , Isoenzymes/genetics , Isoenzymes/metabolism , Lysosomes/drug effects , Osteoclasts/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , Proto-Oncogene Proteins c-raf/drug effects , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-yes , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Tartrate-Resistant Acid Phosphatase , Transfection
10.
J Cancer Res Clin Oncol ; 126(11): 631-40, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11079727

ABSTRACT

PURPOSE: The purpose of this study was to elucidate the potential of human breast cancer cells (BCC) to induce matrix degradation and neo-vascularization, essential for continued tumor growth, in osteolytic lesions. METHODS: BCC were inoculated into the left cardiac ventricle of female athymic mice and osteolytic lesions were radiologically visualized within 4 weeks from inoculation. RESULTS: Histomorphometric analysis of bone sections revealed a significant increase in the number and maturity of osteoclasts (OCl) lining the bone surfaces next to tumor tissue when compared to corresponding bone surfaces in healthy mice. In addition, a large number of newly formed blood vessels could be visualized by immunohistochemistry at the periphery of and within tumor tissue. When bone marrow (BM) cells were cultured in the presence of BCC the OCl formation was increased threefold. These OCl were also found to be more mature and to have greater resorptive activity. Moreover, BCC were found to stimulate proliferation, migration, and differentiation of BM-derived endothelial cells. CONCLUSIONS: Matrix destruction and neo-vascularization are accomplished by BCC arrested in the BM cavity by increasing recruitment and activity of OCl and by induction of angiogenesis within or in proximity to the tumor tissue.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Communication/physiology , Neovascularization, Pathologic/pathology , Osteoclasts/pathology , Animals , Bone Neoplasms/blood supply , Bone Neoplasms/pathology , Bone Resorption/etiology , Bone Resorption/pathology , Breast Neoplasms/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cell Movement/physiology , Coculture Techniques , Cytokines/biosynthesis , Cytokines/physiology , Endothelium, Vascular/pathology , Extracellular Matrix/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/etiology , Osteolysis , Tumor Cells, Cultured
11.
Cancer Res ; 59(22): 5849-55, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10582709

ABSTRACT

Metastatic breast cancer causes destruction of significant amounts of bone, and, although bone is the most likely site of breast cancer metastasis, little is understood about interactions between tumor cells and bone-resorbing osteoclasts. We have investigated the paracrine factors produced by breast cancer cells that are involved in increasing osteoclast activity. We have determined by immunoassay that the human breast cancer cell line MDA MB 231 (231) cultured in serum-free medium secretes transforming growth factors type beta(TGF-beta) 1 and 2, macrophage colony-stimulating factor (M-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), interleukin (IL) -1 and -6, tumor necrosis factor alpha (TNF-alpha), insulin-like growth factor II (IGF II), and parathyroid hormone-related peptide. To determine which of these are involved in increased bone destruction, we have fractionated serum-free 231-conditioned media and measured these fractions for effects on osteoclast resorption activity using multiple activity assays. The pattern of responses was complex. Several fractions stimulated osteoclast resorption either by increasing the number of osteoclasts binding to the bone or by elevating the resorption activity of the individual osteoclasts. Other fractions inhibited osteoclast activity. Analysis of active fractions for the factors identified in the 231-conditioned medium revealed that the presence of TNF-alpha and IGF-II was restricted to separate fractions that stimulated osteoclast resorption activity. The fractions that inhibited osteoclast resorption activity contained M-CSF, IL-6, TGF-beta2, and GM-CSF. No TGF-beta1 or IL-1 was detected in any of the active fractions. Our data support the hypothesis that breast cancer cells modulate osteoclast activity using multiple regulatory factors that increase both the number of mature osteoclasts attached to the bone and the bone resorption activity of these individual osteoclasts. Once it is understood how metastatic breast cancer elevates osteoclast-mediated bone loss, effective therapies to slow the progression and/or prevent this bone loss will become possible.


Subject(s)
Bone Resorption/etiology , Breast Neoplasms/chemistry , Cytokines/analysis , Neoplasm Proteins/analysis , Osteoclasts/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chickens , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Cytokines/pharmacology , Female , Growth Substances/analysis , Growth Substances/pharmacology , Humans , Neoplasm Proteins/pharmacology , Tumor Cells, Cultured
12.
Proc Natl Acad Sci U S A ; 96(2): 505-10, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892663

ABSTRACT

For many years it has been recognized that sex steroids have profound effects on bone metabolism. The current perception is that estrogen decreases bone resorption and androgen increases bone deposition. To investigate the potential for androgens to directly modulate bone resorption, we have examined avian osteoclast and human and mouse osteoclast-like cells for androgen responsiveness. There was a dose-dependent decrease in resorption activity in response to alpha-dihydrotestosterone (alpha-DHT), beta-DHT, testosterone, or the synthetic androgen RU1881. This decrease was blocked by cotreatment with the specific androgen antagonist hydroxyflutamide. Further examination of avian osteoclasts revealed that the cells exhibited specific and saturable nuclear binding of tritiated RU1881 and that alpha-DHT stimulated the activity of the androgen response element as measured by using a chloramphenicol acetyltransferase reporter plasmid. In addition, avian osteoclasts responded to androgen treatment with elevated production and secretion of transforming growth factor beta, a well documented response to androgen exposure in other cell systems. Treatment with either alpha-DHT or beta-DHT for 24 hours resulted in a significant dose-dependent decrease in secretion of cathepsin B and tartrate-resistant acid phosphatase. This response to beta-DHT, a stereoisomer of alpha-DHT that is inactive in other androgen receptor-dependent systems, supports the hypothesis that the osteoclast androgen receptor has unusual ligand-binding properties. Taken together, these results confirm the presence of functional androgen receptors in these cells and support the conclusion that osteoclasts are able to respond directly to androgens in vitro and thus are potential androgen target cells in vivo.


Subject(s)
Androgens/pharmacology , Bone Resorption/metabolism , Osteoclasts/metabolism , Acid Phosphatase/metabolism , Androgen Antagonists/pharmacology , Animals , Cathepsin B/metabolism , Chickens , Dihydrotestosterone/pharmacology , Flutamide/analogs & derivatives , Flutamide/pharmacology , Genes, Reporter/drug effects , Genes, Reporter/genetics , Humans , Isoenzymes/metabolism , Metribolone/pharmacology , Mice , Receptors, Androgen/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Tartrate-Resistant Acid Phosphatase , Transfection/genetics , Transforming Growth Factor beta/metabolism
13.
Crit Rev Eukaryot Gene Expr ; 8(2): 125-40, 1998.
Article in English | MEDLINE | ID: mdl-9714894

ABSTRACT

Estrogen has beneficial effects on the maintenance of bone mass and cardiovascular health; however, these are counterbalanced by the potential detrimental effects in reproductive tissues. The paradox of estrogen and bone metabolism is highlighted by the observation that estrogen analogs can act as estrogen antagonists in reproductive tissue while acting as agonists in bone. Estrogen effects on osteoblast and osteoclast gene expression are of critical importance in seeking to understand these contradictory influences. In this review, the in vivo evidence that drives the need for discovery of estrogen effects on bone cells is briefly discussed. In order to better fit this discussion of estrogen actions into a broader context of steroid action, there is a brief review of the cascade model for steroid hormone action and the growing area of nongenomic actions of steroids. This is followed by a more detailed examination of our present knowledge of estrogen effects on osteoblasts and the differentiation and activity of osteoclasts. Direct effects of estrogen on osteoblast-like cell proliferation, cytokine expression, and responses to other stimuli are addressed. The effects of estrogen on cytokine modulation of osteoclastogenesis is examined as well as direct actions of estrogen on osteoclast activity and programmed cell death.


Subject(s)
Estrogens/physiology , Gene Expression Regulation/physiology , Osteoblasts/metabolism , Osteoclasts/metabolism , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Humans , Osteoblasts/cytology , Osteoclasts/cytology
14.
J Bone Miner Res ; 13(1): 67-78, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443792

ABSTRACT

Osteoclasts generate high levels of superoxide anions during bone resorption that contribute to the degradative process, although excessive levels of this free radical may be damaging. One mechanism for their removal is via superoxide dismutase (SOD), a protective superoxide scavenging enzyme. We have previously described a novel developmentally regulated 150 kDa plasma membrane glycoprotein of avian osteoclasts which is reactive with the osteoclast-specific monoclonal antibody (Mab) 121F and is related immunologically, biochemically, and in protein sequence to mitochondrial Mn2+/Fe2+ SOD. We hypothesized that this unusual osteoclast surface component may be involved in protection against superoxides generated during active bone resorption. Increasing concentrations of monovalent Fab fragments prepared from Mab 121F, but not those from another antiosteoclast Mab designated 29C, markedly inhibited both bone particle and bone pit resorption by avian osteoclasts, while reducing tartrate-resistant acid phosphatase activity and causing the morphological contraction of osteoclasts on bone. Thus, the SOD-related membrane antigen may be essential for osteoclast bone resorption. Osteoclast superoxide production, monitored kinetically by cytochrome c reduction and histochemically by nitroblue tetrazolium reduction staining, was significantly greater in the presence of 121F, but not 29C, Fab treatment. Furthermore, the release of another free radical known as nitric oxide, which is produced by osteoclasts, can scavenge superoxides, and acts to potently inhibit osteoclast bone resorption, was dose-dependently increased by 121F Fab in resorbing osteoclast cultures. Therefore, Mab 121F binding may block the potential protective function of the osteoclast plasma membrane SOD-related glycoprotein, leading to a rapid elevation of superoxide levels and a subsequent rise in osteoclast nitric oxide release, feedback messages which may be sensed by the osteoclast as signals to cease active bone resorption.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Resorption/immunology , Bone Resorption/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , Acid Phosphatase/immunology , Acid Phosphatase/metabolism , Animals , Bone Resorption/pathology , Cell Size/immunology , Cells, Cultured , Chickens , Enzyme Activation/immunology , Humerus , Isoenzymes/immunology , Isoenzymes/metabolism , Nitric Oxide/metabolism , Osteoclasts/ultrastructure , Superoxides/metabolism , Tartrate-Resistant Acid Phosphatase , Tibia
16.
J Bone Miner Res ; 12(5): 742-52, 1997 May.
Article in English | MEDLINE | ID: mdl-9144340

ABSTRACT

Isolated osteoclasts from 5-week-old chickens respond to estradiol treatment in vitro with decreased resorption activity, increased nuclear proto-oncogene expression, and decreased lysosomal enzyme secretion. This study examines osteoclasts from embryonic chickens and egg-laying hens for evidence of estrogen responsiveness. Although osteoclasts from both of these sources express estrogen receptor mRNA and protein, estradiol treatment had no effect on resorption activity. In contrast to the lack of effect on resorption, estradiol treatment for 30 minutes resulted in steady-state mRNA levels of c-fos and c-jun increasing in osteoclasts from embryonic chickens and decreasing in osteoclasts from egg-laying hens. These data suggest that a nuclear proto-oncogene response may not be involved in estradiol-mediated decreased osteoclast resorption activity. To examine the influence of circulating estrogen on osteoclast estrogen responsiveness, 5-week-old chickens were injected with estrogen for 4 days prior to sacrifice. Estradiol treatment of osteoclasts from these chickens did not decrease resorption activity in vitro. Transfection of an estrogen receptor expression vector into osteoclasts from the estradiol-injected chickens and egg-laying hens restored estrogen responsiveness. Osteoclasts from 5-week-old chickens and estradiol treated 5-week-old chickens transfected with the estrogen receptor expression vector contained significantly higher levels of estrogen receptor protein and responded to estradiol treatment by decreasing secretion of cathepsins B and L and tartrate-resistant acid phosphatase. In contrast, osteoclasts from embryonic chickens, egg-laying hens, and estradiol-treated 5-week-old chickens either untransfected or transfected with an empty expression vector did not respond similarly. These data suggest that modulation of osteoclast estrogen responsiveness may be controlled by changes in the osteoclast estrogen receptor levels.


Subject(s)
Aging/metabolism , Estradiol/pharmacology , Osteoclasts/drug effects , Receptors, Estrogen/metabolism , Animals , Chick Embryo , Chickens , Drug Evaluation, Preclinical , Female , In Vitro Techniques
17.
J Orthop Res ; 14(3): 396-402, 1996 May.
Article in English | MEDLINE | ID: mdl-8676252

ABSTRACT

The cellular mechanism through which osseous breast cancer metastases induce the focal destruction of bone (tumor osteolysis) is unknown. An athymic mouse model designed for the study of tumor osteolysis was developed and the influence of two human breast cancer tumors on bone was studied. Tumor-induced osteolysis occurred between 7 and 10 weeks after inoculation of mouse femora with MDA-MB-231 or MDA-MB-435s breast cancer cells. An increase in osteoclast number and an increase in osteoclast size (area) were detected when tumor-bearing and sham-injected limbs were compared. In vitro analysis of the influence of the tumor-conditioned medium on osteoclast-mediated bone resorption revealed that this conditioned medium stimulated the resorption by increasing both the number of osteoclasts bound to bone and the number of bone resorption pits formed per osteoclast. In addition, in vitro analysis of the influence of breast cancer tumor cells on osteoclast formation or survival, or both, demonstrated that breast cancer cells induced a dramatic increase in the number of osteoclasts detected in culture. Taken in total these findings suggest that human breast cancer tumors induce osteolysis by enhancing osteoclast adherence to bone, stimulating osteoclast-mediated bone resorption and either prolonging the survival of osteoclasts or increasing osteoclast formation.


Subject(s)
Bone Neoplasms/secondary , Osteoclasts/cytology , Osteolysis/pathology , Animals , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/pathology , Breast Neoplasms , Cell Count , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured/cytology , Cellular Senescence/physiology , Culture Media, Conditioned , Female , Femur/diagnostic imaging , Femur/pathology , Humans , Mice , Mice, Nude , Radiography , Tumor Cells, Cultured/cytology
18.
Endocrinology ; 137(2): 615-21, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8593810

ABSTRACT

Estrogen deficiency induced by menopause leads to an increase in bone resorption that is not compensated for by a comparable increase in bone formation, resulting in excessive bone loss. Clinically, estrogen replacement reverses these processes, but the mechanisms by which this takes place are not completely understood. Both osteoclasts and osteoblasts contain functional estrogen receptors and, therefore, may be directly involved in these responses. Because both osteoclasts and osteoblasts secrete transforming growth factor-beta (TGF beta), and because 17 beta-estradiol (E2) treatment increases TGF beta production by osteoblast-like cells in vitro, we have investigated the possibility that E2 also may increase the production of TGF beta by isolated osteoclasts in vitro. Highly purified avian osteoclasts were treated with either vehicle or E2, and TGF beta protein accumulation in culture was measured by bioassay. Although an E2 dose-dependent increase in TGF beta protein accumulation in osteoclast-conditioned medium was measured at 4 h of treatment, a steroid dose-dependent decrease in the accumulation of active TGF beta was measured after 18 h of estrogen treatment. The steroid specificity of the increased TGF beta accumulation was confirmed by demonstrating that the E2-induced increase in TGF beta protein levels in the medium was inhibited by cotreatment with a specific E2 antagonist. Interestingly, E2 treatment induced a TGF beta isoform change from TGF beta 2 to predominantly TGF beta 3. Thus, the data suggest that a direct action of E2 on osteoclasts to lower resorption activity may be mediated by autocrine/paracrine production and activation of TGF beta, perhaps including modulation of specific isoform production.


Subject(s)
Estrogens/pharmacology , Osteoclasts/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Chickens , Dose-Response Relationship, Drug , Isomerism , Osmolar Concentration , Osteoclasts/drug effects , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/genetics
19.
Nucleic Acids Res ; 23(23): 4907-12, 1995 Dec 11.
Article in English | MEDLINE | ID: mdl-8532536

ABSTRACT

The TGF-beta family of growth factors has been extensively studied and found to play major roles in bone physiology and disease. A novel, TGF-beta-inducible early gene (TIEG) in normal human fetal osteoblasts (hFOB) has been identified using differential-display PCR. Using this differentially expressed cDNA fragment of TIEG to screen a hOB cDNA library, a near full-length cDNA for this gene was isolated. Northern analyses indicated that the steady-state levels of the 3.5 kb TIEG mRNA increased within 30 min of TGF-beta treatment of human osteoblasts and reached a maximum of 10-fold above control levels at 120 min post-treatment. This regulation was independent of new protein synthesis. Computer sequence analyses indicates that TIEG mRNA encodes for a 480 amino-acid protein. The TIEG protein contains three zinc finger motifs, several proline-rich src homology-3 (SH3) binding domains at the C-terminal end, and is homologous in this region to the zinc finger-containing transcription factor family of genes. A growth factor/cytokine-specific induction of TIEG has been shown. TIEG expression in hFOB cells was highly induced by TGF-beta and bone morphogenetic protein-2 (BMP-2), with a moderate induction by epidermal growth factor (EGF), but no induction by other growth factors/cytokines was observed. In addition to osteoblastic cells, high levels of TIEG expression were detected in skeletal muscle tissue, while low or no detectable levels were found in brain, lung, liver or kidney. Because TIEG is an early induced putative transcription factor gene, and shows a growth factor induction and tissue specificity, its protein product might play an important role as a signalling molecule in osteoblastic cells.


Subject(s)
Genes, Immediate-Early/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/pharmacology , Zinc Fingers , Base Sequence , Cell Line , Dactinomycin/pharmacology , Genes, Immediate-Early/drug effects , Humans , Molecular Sequence Data , Organ Specificity , Osteoblasts/metabolism , Polymerase Chain Reaction , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/analysis , RNA, Messenger/chemistry , Transforming Growth Factor beta/physiology
20.
J Cell Biochem ; 58(1): 39-46, 1995 May.
Article in English | MEDLINE | ID: mdl-7642721

ABSTRACT

The effects of Zeolite A on bone resorbing activity of highly purified avian osteoclasts were analyzed. The present study demonstrates that when 100 micrograms/ml of acid-treated Zeolite A is added to the media the number of pits per osteoclast is reduced 3-fold at 24 h after treatment. Secreted cathepsin B enzyme activity was also reduced 3-fold. A similar reduction in pit number per osteoclast was measured following 48 h of treatment with Zeolite A but there appeared to be less reduction of cathepsin B enzyme activity. The effects on pit number and cathepsin B protein activity were Zeolite dose dependent. The structure of the compound seemed to be responsible for the effects measured since compounds used to represent constituents of Zeolite A (silicon dioxide and aluminum chloride) failed to inhibit bone resorption or reduce the level of secreted cathepsin B enzyme activity. Thus the molecular architecture of Zeolite A or a derivative thereof appears to be important. In conclusion, the data indicate that Zeolite A can inhibit bone resorption. Together with previous data on osteoblasts, this might suggest a potential positive activity of intact Zeolite A or a partial substructure of Zeolite A on bone turnover.


Subject(s)
Bone Resorption/prevention & control , Zeolites/pharmacology , Aluminum Chloride , Aluminum Compounds/pharmacology , Animals , Bone Resorption/metabolism , Bone and Bones/metabolism , Cathepsin B/metabolism , Chickens , Chlorides/pharmacology , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Osteoclasts/drug effects , Osteoclasts/metabolism , Silicon Dioxide/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...