Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
J Bone Miner Res ; 21(4): 565-75, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16598377

ABSTRACT

UNLABELLED: CP432 is a newly discovered, nonprostanoid EP4 receptor selective prostaglandin E2 agonist. CP432 stimulates trabecular and cortical bone formation and restores bone mass and bone strength in aged ovariectomized rats with established osteopenia. INTRODUCTION: The purpose of this study was to determine whether a newly discovered, nonprostanoid EP4 receptor selective prostaglandin E2 (PGE2) agonist, CP432, could produce bone anabolic effects in aged, ovariectomized (OVX) rats with established osteopenia. MATERIALS AND METHODS: CP432 at 0.3, 1, or 3 mg/kg/day was given for 6 weeks by subcutaneous injection to 12-month-old rats that had been OVX for 8.5 months. The effects on bone mass, bone formation, bone resorption, and bone strength were determined. RESULTS: Total femoral BMD increased significantly in OVX rats treated with CP432 at all doses. CP432 completely restored trabecular bone volume of the third lumbar vertebral body accompanied with a dose-dependent decrease in osteoclast number and osteoclast surface and a dose-dependent increase in mineralizing surface, mineral apposition rate, and bone formation rate-tissue reference in OVX rats. CP432 at 1 and 3 mg/kg/day significantly increased total tissue area, cortical bone area, and periosteal and endocortical bone formation in the tibial shafts compared with both sham and OVX controls. CP432 at all doses significantly and dose-dependently increased ultimate strength in the fifth lumber vertebral body compared with both sham and OVX controls. At 1 and 3 mg/kg/day, CP432 significantly increased maximal load in a three-point bending test of femoral shaft compared with both sham and OVX controls. CONCLUSIONS: CP432 completely restored trabecular and cortical bone mass and strength in established osteopenic, aged OVX rats by stimulating bone formation and inhibiting bone resorption on trabecular and cortical surfaces.


Subject(s)
Aging/physiology , Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Dinoprostone/agonists , Osteogenesis/drug effects , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/metabolism , Animals , Body Weight , Bone Density/physiology , Disease Models, Animal , Female , Femur/anatomy & histology , Lumbar Vertebrae/anatomy & histology , Molecular Structure , Organ Size/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype , Substrate Specificity , Tibia/anatomy & histology
2.
Mol Endocrinol ; 17(7): 1356-67, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12677010

ABSTRACT

The P2X7 nucleotide receptor is an ATP-gated ion channel expressed widely in cells of hematopoietic origin. Our purpose was to explore the involvement of the P2X7 receptor in bone development and remodeling by characterizing the phenotype of mice genetically modified to disrupt the P2X7 receptor [knockout (KO)]. Femoral length did not differ between KO and wild-type (WT) littermates at 2 or 9 months of age, indicating that the P2X7 receptor does not regulate longitudinal bone growth. However, KO mice displayed significant reduction in total and cortical bone content and periosteal circumference in femurs, and reduced periosteal bone formation and increased trabecular bone resorption in tibias. Patch clamp recording confirmed expression of functional P2X7 receptors in osteoclasts from WT but not KO mice. Osteoclasts were present in vivo and formed in cultures of bone marrow from KO mice, indicating that this receptor is not essential for fusion of osteoclast precursors. Functional P2X7 receptors were also found in osteoblasts from WT but not KO mice, suggesting a direct role in bone formation. P2X7 receptor KO mice demonstrate a unique skeletal phenotype that involves deficient periosteal bone formation together with excessive trabecular bone resorption. Thus, the P2X7 receptor represents a novel therapeutic target for the management of skeletal disorders such as osteoporosis.


Subject(s)
Bone Development/genetics , Bone Resorption/genetics , Receptors, Purinergic P2/physiology , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Female , Femur/anatomy & histology , Femur/growth & development , Femur/pathology , Gene Expression Regulation, Developmental , Male , Mice , Mice, Knockout , Osteoblasts/physiology , Osteoclasts/physiology , Patch-Clamp Techniques , Receptors, Purinergic P2X7 , Tomography/methods
3.
BMC Cell Biol ; 3: 15, 2002 Jun 21.
Article in English | MEDLINE | ID: mdl-12097147

ABSTRACT

BACKGROUND: The intracellular signaling events of the bone morphogenetic proteins (BMPs) involve the R-Smad family members Smad1, Smad5, Smad8 and the Co-Smad, Smad4. Smads are currently considered to be DNA-binding transcriptional modulators and shown to recruit the master transcriptional co-activator CBP/p300 for transcriptional activation. SNIP1 is a recently discovered novel repressor of CBP/p300. Currently, the detailed molecular mechanisms that allow R-Smads and Co-Smad to co-operatively modulate transcription events are not fully understood. RESULTS: Here we report a novel physical and functional link between Smad1 and the 26S proteasome that contributes to Smad1- and Smad4-mediated transcriptional regulation. Smad1 forms a complex with a proteasome beta subunit HsN3 and the ornithine decarboxylase antizyme (Az). The interaction is enhanced upon BMP type I receptor activation and occur prior to the incorporation of HsN3 into the mature 20S proteasome. Furthermore, BMPs trigger the translocation of Smad1, HsN3 and Az into the nucleus, where the novel CBP/p300 repressor protein SNIP1 is further recruited to Smad1/HsN3/Az complex and degraded in a Smad1-, Smad4- and Az-dependent fashion. The degradation of the CBP/p300 repressor SNIP1 is likely an essential step for Smad1-, Smad4-mediated transcriptional activation, since increased SNIP1 expression inhibits BMP-induced gene responses. CONCLUSIONS: Our studies thus add two additional important functional partners of Smad1 into the signaling web of BMPs and also suggest a novel mechanism for Smad1 and Smad4 to co-modulate transcription via regulating proteasomal degradation of CBP/p300 repressor SNIP1.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cysteine Endopeptidases/metabolism , Intracellular Signaling Peptides and Proteins , Multienzyme Complexes/metabolism , Signal Transduction , Transforming Growth Factor beta , Animals , Blotting, Western , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cysteine Endopeptidases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Mutation , Precipitin Tests , Proteasome Endopeptidase Complex , Protein Binding/drug effects , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Smad Proteins , Smad1 Protein , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Two-Hybrid System Techniques
4.
Proc Natl Acad Sci U S A ; 104(25): 10619-24, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17537919

ABSTRACT

Bone is accrued and maintained primarily through the coupled actions of bone-forming osteoblasts and bone-resorbing osteoclasts. Cumulative in vitro studies indicated that proline-rich tyrosine kinase 2 (PYK2) is a positive mediator of osteoclast function and activity. However, our investigation of PYK2-/- mice did not reveal evidence supporting an essential function for PYK2 in osteoclasts either in vivo or in culture. We find that PYK2-/- mice have high bone mass resulting from an unexpected increase in bone formation. Consistent with the in vivo findings, mouse bone marrow cultures show that PYK2 deficiency enhances differentiation and activity of osteoprogenitor cells, as does expressing a PYK2-specific short hairpin RNA or dominantly interfering proteins in human mesenchymal stem cells. Furthermore, the daily administration of a small-molecule PYK2 inhibitor increases bone formation and protects against bone loss in ovariectomized rats, an established preclinical model of postmenopausal osteoporosis. In summary, we find that PYK2 regulates the differentiation of early osteoprogenitor cells across species and that inhibitors of the PYK2 have potential as a bone anabolic approach for the treatment of osteoporosis.


Subject(s)
Focal Adhesion Kinase 2/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Osteogenesis/physiology , Osteoporosis/therapy , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Cells, Cultured , Enzyme Inhibitors/therapeutic use , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Knockout , Ovariectomy , Rats , Rats, Sprague-Dawley
5.
Bioorg Med Chem Lett ; 16(7): 1799-802, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16442794

ABSTRACT

Heptanoic acid lactams, exemplified by 2, were identified as highly selective EP4 agonists via high throughput screening. Lead optimization led to the identification of lactams with a 30-fold increase in EP4 potency in vitro. Compounds demonstrated robust bone anabolic effects when administered in vivo in rat models of osteoporosis.


Subject(s)
Bone Density/drug effects , Bone Development , Bone and Bones/drug effects , Organ Size/drug effects , Ovariectomy , Receptors, Prostaglandin E/agonists , Animals , Female , Osteoporosis , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype
SELECTION OF CITATIONS
SEARCH DETAIL