Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Endocrinology ; 142(10): 4295-304, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564687

ABSTRACT

PTH is a potent bone anabolic factor, and its combination with antiresorptive agents has been proposed as a therapy for osteoporosis. We tested the effects of PTH, alone and in combination with the novel antiresorptive agent OPG, in a rat model of severe osteopenia. Sprague Dawley rats were sham-operated or ovariectomized at 3 months of age. Rats were untreated for 15 months, at which time ovariectomy had caused significant decreases in bone mineral density in the lumbar vertebrae and femur. Rats were then treated for 5.5 months with vehicle (PBS), human PTH-(1-34) (80 microg/kg), rat OPG (10 mg/kg), or OPG plus PTH (all three times per wk, sc). Treatment of ovariectomized rats with OPG or PTH alone increased bone mineral density in the lumbar vertebrae and femur, whereas PTH plus OPG caused significantly greater and more rapid increases than either therapy alone (P < 0.05). OPG significantly reduced osteoclast surface in the lumbar vertebrae and femur (P < 0.05 vs. sham or ovariectomized), but had no effect on osteoblast surface at either site. Ovariectomy significantly decreased the mechanical strength of the lumbar vertebrae and femur. In the lumbar vertebrae, OPG plus PTH was significantly more effective than PTH alone at reversing ovariectomy-induced deficits in stiffness and elastic modulus. These data suggest that OPG plus PTH represent a potentially useful therapeutic option for patients with severe osteoporosis.


Subject(s)
Bone Diseases, Metabolic/drug therapy , Glycoproteins/pharmacology , Peptide Fragments/pharmacology , Teriparatide/pharmacology , Animals , Bone Density/drug effects , Bone Diseases, Metabolic/physiopathology , Drug Interactions , Drug Therapy, Combination , Female , Glycoproteins/therapeutic use , Osteoprotegerin , Ovariectomy , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/therapeutic use , Receptors, Tumor Necrosis Factor , Teriparatide/analogs & derivatives , Teriparatide/therapeutic use
2.
Cancer Res ; 61(11): 4432-6, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389072

ABSTRACT

Certain malignancies, including breast cancer, frequently metastasize to bone, where the tumor cells induce osteoclasts to locally destroy bone. Osteoprotegerin (OPG), a member of the tumor necrosis factor receptor family, is a negative regulator of osteoclast differentiation, activation, and survival. We tested the ability of recombinant OPG to inhibit tumor-induced osteoclastogenesis, osteolysis, and skeletal tumor burden in two animal models. In a syngeneic model, mouse colon adenocarcinoma (Colon-26) cells were injected into the left ventricle of mice. Treatment with OPG dose-dependently decreased the number and area of radiographically evident lytic bone lesions, which, at the highest dose, were undetectable. Histologically, OPG also decreased skeletal tumor burden and tumor-associated osteoclasts. In a nude mouse model, OPG treatment completely prevented radiographic osteolytic lesions caused by human MDA-MB-231 breast cancer cells. Histologically, OPG decreased skeletal tumor burden by 75% and completely eradicated MDA tumor-associated osteoclasts. In both models, OPG had no effect on metastatic tumor burden in a panel of soft tissue organs. These data indicate that OPG may be an effective therapy for preventing osteolysis and decreasing skeletal tumor burden in patients with bone metastasis.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Glycoproteins/pharmacology , Osteolysis/drug therapy , Adenocarcinoma/pathology , Animals , Bone Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Transformed , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Nude , Osteoprotegerin , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor , Xenograft Model Antitumor Assays
3.
Mol Ther ; 3(2): 197-205, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11237676

ABSTRACT

Osteoprotegerin (OPG) regulates bone resorption by inhibiting osteoclast formation, function, and survival. The current studies employed a mouse ovariectomy (OVX) model of estrogen deficiency to investigate gene therapy with OPG as a means of preventing osteoporosis. Young adult females injected with a recombinant adenoviral (Ad) vector carrying cDNA of either full-length OPG or a fusion protein combining the hOPG ligand-binding domain with the human immunoglobulin constant domain (Ad-hOPG-Fc) developed serum OPG concentrations exceeding the threshold needed for efficacy. However, elevated circulating OPG levels were sustained for up to 18 months only in mice given Ad-hOPG-Fc. Administration of Ad-hOPG-Fc titers between 10(7) and 10(9) pfu yielded dose-dependent increases in serum OPG. Mice subjected to OVX or sham surgery followed by immediate treatment with Ad-hOPG-Fc had significantly more bone volume with reduced osteoclast numbers in axial and appendicular bones after 4 weeks. In contrast, animals given OVX and either a control vector or vehicle had significantly less bone than did comparably treated sham-operated mice. This study demonstrates that a single adenoviral gene transfer can produce persistent high-level OPG expression and shows that gene therapy to provide sustained delivery of OPG may prove useful in treating osteoporosis.


Subject(s)
Adenoviridae/genetics , Glycoproteins/genetics , Osteoporosis/therapy , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Biological Assay , Blotting, Southern , Blotting, Western , Bone Density/drug effects , Bone Resorption , DNA, Complementary/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Humans , Ligands , Mice , Mice, Inbred C57BL , Osteoprotegerin , Ovariectomy , Ovary/physiology , Pelvis/diagnostic imaging , Radiography , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/metabolism , Time Factors
4.
J Exp Med ; 192(4): 463-74, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10952716

ABSTRACT

High systemic levels of osteoprotegerin (OPG) in OPG transgenic mice cause osteopetrosis with normal tooth eruption and bone elongation and inhibit the development and activity of endosteal, but not periosteal, osteoclasts. We demonstrate that both intravenous injection of recombinant OPG protein and transgenic overexpression of OPG in OPG(-/-) mice effectively rescue the osteoporotic bone phenotype observed in OPG-deficient mice. However, intravenous injection of recombinant OPG over a 4-wk period could not reverse the arterial calcification observed in OPG(-/-) mice. In contrast, transgenic OPG delivered from mid-gestation through adulthood does prevent the formation of arterial calcification in OPG(-/-) mice. Although OPG is normally expressed in arteries, OPG ligand (OPGL) and receptor activator of NF-kappaB (RANK) are not detected in the arterial walls of wild-type adult mice. Interestingly, OPGL and RANK transcripts are detected in the calcified arteries of OPG(-/-) mice. Furthermore, RANK transcript expression coincides with the presence of multinuclear osteoclast-like cells. These findings indicate that the OPG/OPGL/RANK signaling pathway may play an important role in both pathological and physiological calcification processes. Such findings may also explain the observed high clinical incidence of vascular calcification in the osteoporotic patient population.


Subject(s)
Bone Density/physiology , Calcinosis/physiopathology , Glycoproteins/metabolism , Osteoclasts/metabolism , Osteopetrosis/metabolism , Osteoporosis/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Acid Phosphatase/metabolism , Animals , Aorta/pathology , Blotting, Western , CHO Cells , Cathepsin K , Cathepsins/metabolism , Cricetinae , Femur/anatomy & histology , Femur/diagnostic imaging , Femur/metabolism , Glycoproteins/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Isoenzymes/metabolism , Mice , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoclasts/ultrastructure , Osteopetrosis/genetics , Osteoporosis/genetics , Osteoprotegerin , Radiography , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/metabolism , Tartrate-Resistant Acid Phosphatase
5.
Proc Natl Acad Sci U S A ; 97(4): 1566-71, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677500

ABSTRACT

We have generated RANK (receptor activator of NF-kappaB) nullizygous mice to determine the molecular genetic interactions between osteoprotegerin, osteoprotegerin ligand, and RANK during bone resorption and remodeling processes. RANK(-/-) mice lack osteoclasts and have a profound defect in bone resorption and remodeling and in the development of the cartilaginous growth plates of endochondral bone. The osteopetrosis observed in these mice can be reversed by transplantation of bone marrow from rag1(-/-) (recombinase activating gene 1) mice, indicating that RANK(-/-) mice have an intrinsic defect in osteoclast function. Calciotropic hormones and proresorptive cytokines that are known to induce bone resorption in mice and human were administered to RANK(-/-) mice without inducing hypercalcemia, although tumor necrosis factor alpha treatment leads to the rare appearance of osteoclast-like cells near the site of injection. Osteoclastogenesis can be initiated in RANK(-/-) mice by transfer of the RANK cDNA back into hematopoietic precursors, suggesting a means to critically evaluate RANK structural features required for bone resorption. Together these data indicate that RANK is the intrinsic cell surface determinant that mediates osteoprotegerin ligand effects on bone resorption and remodeling as well as the physiological and pathological effects of calciotropic hormones and proresorptive cytokines.


Subject(s)
Bone and Bones/metabolism , Calcium/metabolism , Carrier Proteins/genetics , Membrane Glycoproteins/genetics , Osteogenesis , Animals , Bone Remodeling/genetics , Bone Resorption/genetics , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Differentiation/genetics , Cytokines/pharmacology , Gene Targeting , Gene Transfer Techniques , Growth Plate/pathology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Knockout , Osteoclasts/pathology , Osteopetrosis/genetics , Osteopetrosis/pathology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Retroviridae/genetics
6.
Cancer Res ; 60(4): 783-7, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706080

ABSTRACT

Osteoprotegerin (OPG), a novel, secreted tumor necrosis factor receptor family member that inhibits osteoclast formation and activity was examined for its activity in a syngeneic tumor model of humoral hypercalcemia of malignancy. Normal mice bearing Colon-26 tumors develop increases in both parathyroid hormone-related protein (PTHrP) expression and plasma PTHrP, marked hypercalcemia, and increased bone resorption. OPG, given either at the onset of hypercalcemia or after it had occurred, blocked tumor-induced increases in bone resorption and hypercalcemia and rapidly normalized blood ionized calcium. In tumor-bearing mice, OPG treatments reduced osteoclast activity from approximately 2-fold above normal into the subphysiological range but had no effects on tumor size, tumor-induced cachexia, or PTHrP levels. The potent effects of OPG in this humoral hypercalcemia of malignancy model suggest a potential therapeutic role for OPG in the prevention and treatment of this disorder.


Subject(s)
Glycoproteins/therapeutic use , Hypercalcemia/prevention & control , Neoplasms, Experimental/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor/therapeutic use , Animals , Dose-Response Relationship, Drug , Hypercalcemia/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Osteoclasts/drug effects , Osteoprotegerin , Parathyroid Hormone-Related Protein , Proteins/analysis
7.
Nature ; 402(6759): 304-9, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10580503

ABSTRACT

Bone remodelling and bone loss are controlled by a balance between the tumour necrosis factor family molecule osteoprotegerin ligand (OPGL) and its decoy receptor osteoprotegerin (OPG). In addition, OPGL regulates lymph node organogenesis, lymphocyte development and interactions between T cells and dendritic cells in the immune system. The OPGL receptor, RANK, is expressed on chondrocytes, osteoclast precursors and mature osteoclasts. OPGL expression in T cells is induced by antigen receptor engagement, which suggests that activated T cells may influence bone metabolism through OPGL and RANK. Here we report that activated T cells can directly trigger osteoclastogenesis through OPGL. Systemic activation of T cells in vivo leads to an OPGL-mediated increase in osteoclastogenesis and bone loss. In a T-cell-dependent model of rat adjuvant arthritis characterized by severe joint inflammation, bone and cartilage destruction and crippling, blocking of OPGL through osteoprotegerin treatment at the onset of disease prevents bone and cartilage destruction but not inflammation. These results show that both systemic and local T-cell activation can lead to OPGL production and subsequent bone loss, and they provide a novel paradigm for T cells as regulators of bone physiology.


Subject(s)
Arthritis, Experimental/immunology , Bone Resorption , Carrier Proteins/physiology , Glycoproteins/physiology , Membrane Glycoproteins/physiology , Receptors, Cytoplasmic and Nuclear , T-Lymphocytes/physiology , Animals , Arthritis, Experimental/pathology , Bone and Bones/pathology , Cartilage/pathology , Cells, Cultured , Coculture Techniques , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Osteoprotegerin , RANK Ligand , Rats , Rats, Inbred Lew , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor
8.
J Bone Miner Res ; 14(9): 1478-85, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10469275

ABSTRACT

Osteoprotegerin (OPG) is a secreted protein that inhibits osteoclast formation and activity and appears to be a critical regulator of bone mass and metabolism. In the current study, mice were challenged with various cytokines and hormones (interleukin-1beta, tumor necrosis factor-alpha, parathyroid hormone, parathyroid hormone-related protein, and 1alpha,25-dihydroxyvitamin D3) that are known to increase bone resorption and cause hypercalcemia and treated concurrently with either a recombinant chimeric Fc fusion form of human OPG, with enhanced biological activity (cOPG) (2.5 mg/kg/day) or vehicle. Mice receiving these bone-resorbing factors became hypercalcemic by day 3 after commencing treatment and had increased bone resorption as evidenced by elevated osteoclast numbers on day 5. Concurrent cOPG treatment prevented hypercalcemia (p < 0.05) and maintained osteoclast numbers in the normal range (p < 0.001). The demonstration that cOPG can inhibit bone resorption suggests that this molecule may be useful in the treatment of diseases including hyperparathyroidism, humoral hypercalcemia of malignancy, osteoporosis, and inflammatory bone disease, which are characterized, in part, by increases in osteoclastic bone resorption.


Subject(s)
Bone Resorption , Calcitriol/pharmacology , Glycoproteins/pharmacology , Hypercalcemia/prevention & control , Interleukin-1/pharmacology , Parathyroid Hormone/pharmacology , Proteins/pharmacology , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Calcium/blood , Cell Count/drug effects , Glycoproteins/chemistry , Humans , Isomerism , Male , Mice , Osteoclasts/drug effects , Osteoprotegerin , Parathyroid Hormone/blood , Parathyroid Hormone-Related Protein , Radiography , Weight Loss/drug effects
9.
J Cell Biol ; 145(3): 527-38, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10225954

ABSTRACT

Osteoprotegerin (OPG) and OPG-ligand (OPGL) potently inhibit and stimulate, respectively, osteoclast differentiation (Simonet, W.S., D.L. Lacey, C.R. Dunstan, M. Kelley, M.-S. Chang, R. Luethy, H.Q. Nguyen, S. Wooden, L. Bennett, T. Boone, et al. 1997. Cell. 89:309-319; Lacey, D.L., E. Timms, H.-L. Tan, M.J. Kelley, C.R. Dunstan, T. Burgess, R. Elliott, A. Colombero, G. Elliott, S. Scully, et al. 1998. Cell. 93: 165-176), but their effects on mature osteoclasts are not well understood. Using primary cultures of rat osteoclasts on bone slices, we find that OPGL causes approximately sevenfold increase in total bone surface erosion. By scanning electron microscopy, OPGL-treated osteoclasts generate more clusters of lacunae on bone suggesting that multiple, spatially associated cycles of resorption have occurred. However, the size of individual resorption events are unchanged by OPGL treatment. Mechanistically, OPGL binds specifically to mature OCs and rapidly (within 30 min) induces actin ring formation; a marked cytoskeletal rearrangement that necessarily precedes bone resorption. Furthermore, we show that antibodies raised against the OPGL receptor, RANK, also induce actin ring formation. OPGL-treated mice exhibit increases in blood ionized Ca++ within 1 h after injections, consistent with immediate OC activation in vivo. Finally, we find that OPG blocks OPGL's effects on both actin ring formation and bone resorption. Together, these findings indicate that, in addition to their effects on OC precursors, OPGL and OPG have profound and direct effects on mature OCs and indicate that the OC receptor, RANK, mediates OPGL's effects.


Subject(s)
Bone Resorption/metabolism , Carrier Proteins/pharmacology , Glycoproteins/pharmacology , Membrane Glycoproteins/pharmacology , Osteoclasts/drug effects , Receptors, Cytoplasmic and Nuclear , Actins/metabolism , Animals , Bone and Bones/chemistry , Bone and Bones/cytology , Bone and Bones/metabolism , Carrier Proteins/metabolism , Cattle , Cell Count , Cell Differentiation/drug effects , Cells, Cultured , Cytoskeleton/metabolism , Glycoproteins/metabolism , Hematopoietic Stem Cells/cytology , Hypercalcemia/chemically induced , Hypercalcemia/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Microscopy, Electron, Scanning , Osteoclasts/cytology , Osteoclasts/ultrastructure , Osteoporosis/metabolism , Osteoprotegerin , RANK Ligand , Rats , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor/metabolism
10.
Genes Dev ; 13(8): 1015-24, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10215628

ABSTRACT

Bone resorption and remodeling is an intricately controlled, physiological process that requires the function of osteoclasts. The processes governing both the differentiation and activation of osteoclasts involve signals induced by osteoprotegerin ligand (OPGL), a member of tumor necrosis factor (TNF) superfamily, and its cognate receptor RANK. The molecular mechanisms of the intracellular signal transduction remain to be elucidated. Here we report that mice deficient in TNF receptor-associated factor 6 (TRAF6) are osteopetrotic with defects in bone remodeling and tooth eruption due to impaired osteoclast function. Using in vitro assays, we demonstrate that TRAF6 is crucial not only in IL-1 and CD40 signaling but also, surprisingly, in LPS signaling. Furthermore, like TRAF2 and TRAF3, TRAF6 is essential for perinatal and postnatal survival. These findings establish unexpectedly diverse and critical roles for TRAF6 in perinatal and postnatal survival, bone metabolism, LPS, and cytokine signaling.


Subject(s)
CD40 Antigens/metabolism , Interleukin-1/metabolism , Lipopolysaccharides/metabolism , Mitogen-Activated Protein Kinases , Osteopetrosis/physiopathology , Proteins/physiology , Signal Transduction , Animals , B-Lymphocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Enzyme Activation , Female , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Mice , Mice, Knockout , NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Proteins/genetics , TNF Receptor-Associated Factor 6
11.
Proc Natl Acad Sci U S A ; 96(7): 3540-5, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097072

ABSTRACT

A receptor that mediates osteoprotegerin ligand (OPGL)-induced osteoclast differentiation and activation has been identified via genomic analysis of a primary osteoclast precursor cell cDNA library and is identical to the tumor necrosis factor receptor (TNFR) family member RANK. The RANK mRNA was highly expressed by isolated bone marrow-derived osteoclast progenitors and by mature osteoclasts in vivo. Recombinant OPGL binds specifically to RANK expressed by transfected cell lines and purified osteoclast progenitors. Transgenic mice expressing a soluble RANK-Fc fusion protein have severe osteopetrosis because of a reduction in osteoclasts, similar to OPG transgenic mice. Recombinant RANK-Fc binds with high affinity to OPGL in vitro and blocks osteoclast differentiation and activation in vitro and in vivo. Furthermore, polyclonal Ab against the RANK extracellular domain promotes osteoclastogenesis in bone marrow cultures suggesting that RANK activation mediates the effects of OPGL on the osteoclast pathway. These data indicate that OPGL-induced osteoclastogenesis is directly mediated through RANK on osteoclast precursor cells.


Subject(s)
Carrier Proteins , Gene Expression Regulation, Developmental , Glycoproteins/pharmacology , Membrane Glycoproteins , Mitogen-Activated Protein Kinases , Osteoclasts/cytology , Receptors, Cytoplasmic and Nuclear , Receptors, Tumor Necrosis Factor/physiology , Animals , Bone Development , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation/drug effects , Cell Line , Cloning, Molecular , Glycoproteins/physiology , Humans , Immunoglobulin G , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Osteoclasts/drug effects , Osteogenesis , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Transcription, Genetic , Transfection
12.
Nature ; 397(6717): 315-23, 1999 Jan 28.
Article in English | MEDLINE | ID: mdl-9950424

ABSTRACT

The tumour-necrosis-factor-family molecule osteoprotegerin ligand (OPGL; also known as TRANCE, RANKL and ODF) has been identified as a potential osteoclast differentiation factor and regulator of interactions between T cells and dendritic cells in vitro. Mice with a disrupted opgl gene show severe osteopetrosis and a defect in tooth eruption, and completely lack osteoclasts as a result of an inability of osteoblasts to support osteoclastogenesis. Although dendritic cells appear normal, opgl-deficient mice exhibit defects in early differentiation of T and B lymphocytes. Surprisingly, opgl-deficient mice lack all lymph nodes but have normal splenic structure and Peyer's patches. Thus OPGL is a new regulator of lymph-node organogenesis and lymphocyte development and is an essential osteoclast differentiation factor in vivo.


Subject(s)
Carrier Proteins , Cytokines/physiology , Embryonic and Fetal Development/physiology , Growth Substances/physiology , Lymph Nodes/embryology , Lymphocytes/cytology , Membrane Glycoproteins , Osteoclasts/cytology , Osteogenesis/physiology , Animals , B-Lymphocytes/cytology , Bone Remodeling/physiology , Cell Differentiation/physiology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Dendritic Cells/cytology , Female , Gene Targeting , Growth Substances/genetics , Hematopoiesis, Extramedullary , Hematopoietic Stem Cells/cytology , Leukopoiesis/physiology , Lymph Nodes/abnormalities , Lymphocyte Activation , Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Mutagenesis , Osteopetrosis/genetics , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/embryology
13.
Genes Dev ; 12(9): 1260-8, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9573043

ABSTRACT

Osteoprotegerin (OPG) is a secreted protein that inhibits osteoclast formation. In this study the physiological role of OPG is investigated by generating OPG-deficient mice. Adolescent and adult OPG-/- mice exhibit a decrease in total bone density characterized by severe trabecular and cortical bone porosity, marked thinning of the parietal bones of the skull, and a high incidence of fractures. These findings demonstrate that OPG is a critical regulator of postnatal bone mass. Unexpectedly, OPG-deficient mice also exhibit medial calcification of the aorta and renal arteries, suggesting that regulation of OPG, its signaling pathway, or its ligand(s) may play a role in the long observed association between osteoporosis and vascular calcification.


Subject(s)
Calcinosis/etiology , Glycoproteins/deficiency , Osteoporosis/etiology , Receptors, Cytoplasmic and Nuclear , Vascular Diseases/etiology , Animals , Arteries/pathology , Bone Density/genetics , Bone Density/physiology , Calcinosis/pathology , Disease Models, Animal , Female , Gene Targeting , Glycoproteins/genetics , Glycoproteins/physiology , In Situ Hybridization , Male , Mice , Mice, Knockout , Osteoporosis/pathology , Osteoprotegerin , Receptors, Tumor Necrosis Factor , Vascular Diseases/pathology
14.
Cell ; 93(2): 165-76, 1998 Apr 17.
Article in English | MEDLINE | ID: mdl-9568710

ABSTRACT

The ligand for osteoprotegerin has been identified, and it is a TNF-related cytokine that replaces the requirement for stromal cells, vitamin D3, and glucocorticoids in the coculture model of in vitro osteoclastogenesis. OPG ligand (OPGL) binds to a unique hematopoeitic progenitor cell that is committed to the osteoclast lineage and stimulates the rapid induction of genes that typify osteoclast development. OPGL directly activates isolated mature osteoclasts in vitro, and short-term administration into normal adult mice results in osteoclast activation associated with systemic hypercalcemia. These data suggest that OPGL is an osteoclast differentiation and activation factor. The effects of OPGL are blocked in vitro and in vivo by OPG, suggesting that OPGL and OPG are key extracellular regulators of osteoclast development.


Subject(s)
Carrier Proteins , Cytokines/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins , Osteoclasts/cytology , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear , Amino Acid Sequence , Animals , Bone Resorption , Cell Differentiation , Cells, Cultured , Cloning, Molecular , Coculture Techniques , Cytokines/genetics , Cytokines/pharmacology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Humans , Hypercalcemia , Ligands , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Molecular Sequence Data , Organ Specificity , Osteoprotegerin , Protein Binding , RANK Ligand , RNA, Messenger/analysis , Rats , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins
15.
J Cell Biochem ; 68(1): 31-49, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9407312

ABSTRACT

We present evidence of cell death by apoptosis during the development of bone-like tissue formation in vitro. Fetal rat calvaria-derived osteoblasts differentiate in vitro, progressing through three stages of maturation: a proliferation period, a matrix maturation period when growth is downregulated and expression of the bone cell phenotype is induced, and a third mineralization stage marked by the expression of bone-specific genes. Here we show for the first time that cells differentiating to the mature bone cell phenotype undergo programmed cell death and express genes regulating apoptosis. Culture conditions that modify expression of the osteoblast phenotype simultaneously modify the incidence of apoptosis. Cell death by apoptosis is directly demonstrated by visualization of degraded DNA into oligonucleosomal fragments after gel electrophoresis. Bcl-XL, an inhibitor of apoptosis, and Bax, which can accelerate apoptosis, are expressed at maximal levels 24 h after initial isolation of the cells and again after day 25 in heavily mineralized bone tissue nodules. Bcl-2 is expressed in a reciprocal manner to its related gene product Bcl-XL with the highest levels observed during the early post-proliferative stages of osteoblast maturation. Expression of p53, c-fos, and the interferon regulatory factors IRF-1 and IRF-2, but not cdc2 or cdk, were also induced in mineralized bone nodules. The upregulation of Msx-2 in association with apoptosis is consistent with its in vivo expression during embryogenesis in areas that will undergo programmed cell death. We propose that cell death by apoptosis is a fundamental component of osteoblast differentiation that contributes to maintaining tissue organization.


Subject(s)
Apoptosis/physiology , CDC2-CDC28 Kinases , Osteoblasts/physiology , Repressor Proteins , Transcription Factors , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fetus/cytology , Fetus/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Osteoblasts/cytology , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Skull/cytology , Skull/physiology
16.
Endocrinology ; 138(5): 2109-16, 1997 May.
Article in English | MEDLINE | ID: mdl-9112411

ABSTRACT

The bone-specific osteocalcin gene is a well established marker of osteoblast activity. We have studied osteocalcin transcription in transgenic mice carrying rat osteocalcin promoter-chloramphenicol acetyltransferase (CAT) reporter constructs. Transgenic lines carrying each of the 1.7-, 1.1-, 0.72-, or 0.35-kilobase promoter constructs expressed the reporter gene in a tissue-specific manner. However, each of these constructs was sensitive to site of integration effects, reflected by a high frequency of nonexpressing transgenic lines. High expression of the 1.7-kilobase promoter in osseous tissues was accompanied by low ectopic expression in the brain. Analysis of CAT expression in femurs, calvariae, and lumbar vertebrae of this line indicated considerable variability in promoter activity among individual transgenic animals. Analysis of the variance in CAT activity demonstrated a linkage between promoter activities in these distant skeletal sites. Promoter activity was inversely correlated with age, and females exhibited severalfold higher activity than age-matched males. Bone marrow stromal cells from these animals, cultured under conditions that support osteoblast differentiation, exhibited the expected postproliferative onset of osteocalcin promoter activity, as assessed by CAT assay. The ex vivo CAT activity was not dependent on the sex or the age of the donor transgenic mouse. Taken together, our results are consistent with the hypothesis that a common, probably humoral, factor(s) regulates osteocalcin transcription in distant skeletal sites. We suggest that the abundance of this factor(s) is different between males and females and among individual mice at a given time point, and that ex vivo culturing of osteoblasts reduces the variation in osteocalcin promoter activity by eliminating the physiological contribution of this factor.


Subject(s)
Bone and Bones/metabolism , Cell Differentiation , Osteoblasts/cytology , Osteocalcin/genetics , Promoter Regions, Genetic , Stromal Cells/cytology , Aging , Animals , Bone Marrow Cells , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , Mice , Mice, Transgenic , Sex Characteristics , Transfection
17.
J Cell Biochem ; 61(1): 152-66, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8726364

ABSTRACT

Fibroblast growth factors (FGF) are osteoblast mitogens, but their effects on bone formation are not clearly understood. Most in vitro studies examining the effects of FGFs on osteoblasts have been performed only during the initial proliferative stage of osteoblast culture. In these studies, we examined the consequential effect of acidic FGF in cultures of rat fetal diploid osteoblasts that undergo a developmental differentiation program producing a mineralized bone-like matrix. During the initial growth period (days 1-10), addition of acidic FGF (100 micrograms/ml) to actively proliferating cells increased (P < 0.05) 3H-thymidine uptake (2,515 +/- 137, mean +/- SEM vs. 5,884 +/- 818 cpm/10(4) cells). During the second stage of maturation (days 10-15), osteoblasts form multilayered nodules of cells and accumulate matrix, followed by mineralization (stage 3, days 16-29). Addition of acidic FGF to the osteoblast cultures from days 7 to 15 completely blocked nodule formation. Furthermore, addition of acidic FGF after nodule formation (days 14-29) inhibited matrix mineralization, which was associated with a marked increase in collagenase gene expression, and resulted in a progressive change in the morphology of the nodules, with only a few remnants of nonmineralized nodules present by day 29. Histochemical and biochemical analyses revealed a decrease in alkaline phosphatase and mineral content, confirming the acidic FGF-induced inhibition of nodule and matrix formation. To identify mechanisms contributing to these changes, we examined expression of cell growth and bone phenotypic markers. Addition of acidic FGF during the proliferative phase (days 7-8) enhanced histone H4, osteopontin, type I collagen, and TGF-beta mRNA levels, which are coupled to proliferating osteoblasts, and blocked the normal developmental increase in alkaline phosphatase and osteocalcin gene expression and calcium accumulation. Addition of acidic FGF to the cultures during matrix maturation (days 14-15) reactivated H4, osteopontin, type I collagen, and TGF-beta gene expression, and decreased alkaline phosphatase and osteocalcin gene expression. In an in vivo experiment, rats were treated with up to 60 micrograms/kg/day acidic FGF intravenously for 30 days. Proliferation of osteoblasts and deposition of bone occurred in the marrow space of the diaphysis of the femur in a dose-related fashion. The metaphyseal areas were unaffected by treatment. In conclusion, our data suggest that acidic FGF is a potent mitogen for early stage osteoblasts which leads to modifications in the formation of the extracellular matrix; increases in TGF-beta and collagenase are functionally implicated in abrogating competency for nodule formation. Persistence of proliferation prevented expression of alkaline phosphatase and osteocalcin, also contributing to the block in the progression of the osteoblast developmental sequence.


Subject(s)
Collagenases/metabolism , Fibroblast Growth Factor 1/pharmacology , Gene Expression Regulation , Osteoblasts/drug effects , Osteoblasts/metabolism , Alkaline Phosphatase/drug effects , Animals , Blotting, Northern , Calcium/analysis , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Collagen/drug effects , Collagenases/genetics , Histones/drug effects , Mitogens , Osteocalcin/drug effects , Osteogenesis/drug effects , Osteopontin , Phosphoproteins/drug effects , Rats , Sialoglycoproteins/drug effects , Time Factors , Transforming Growth Factor beta/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...