Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 148(10): 4984-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17615148

ABSTRACT

Extracellular calcium-sensing receptors (CaRs) and metabotropic or type B gamma-aminobutyric acid receptors (GABA-B-Rs), two closely related members of family C of the G protein-coupled receptor superfamily, dimerize in the formation of signaling and membrane-anchored receptor complexes. We tested whether CaRs and two GABA-B-R subunits (R1 and R2) are expressed in mouse growth plate chondrocytes (GPCs) by PCR and immunocytochemistry and whether interactions between these receptors influence the expression and function of the CaR and extracellular Ca(2+)-mediated cell differentiation. Both CaRs and the GABA-B-R1 and -R2 were expressed in the same zones of the growth plate and extensively colocalized in intracellular compartments and on the membranes of cultured GPCs. The GABA-B-R1 co-immunoprecipitated with the CaR, confirming a physical interaction between the two receptors in GPCs. In vitro knockout of GABA-B-R1 genes, using a Cre-lox recombination strategy, blunted the ability of high extracellular Ca(2+) concentration to activate phospholipase C and ERK1/2, suppressed cell proliferation, and enhanced apoptosis in cultured GPCs. In GPCs, in which the GABA-B-R1 was acutely knocked down, there was reduced expression of early chondrocyte markers, aggrecan and type II collagen, and increased expression of the late differentiation markers, type X collagen and osteopontin. These results support the idea that physical interactions between CaRs and GABA-B-R1s modulate the growth and differentiation of GPCs, potentially by altering the function of CaRs.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/cytology , Chondrocytes/metabolism , Extracellular Fluid/metabolism , Growth Plate/cytology , Receptors, Calcium-Sensing/metabolism , Receptors, GABA-B/physiology , Animals , Apoptosis/physiology , Biomarkers/metabolism , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases , Gene Deletion , Growth Plate/metabolism , Mice , Mice, Inbred BALB C , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/metabolism , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Tissue Distribution , Type C Phospholipases/metabolism
2.
J Am Soc Nephrol ; 18(9): 2509-16, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17656470

ABSTRACT

Thiazide diuretics are used worldwide as a first-choice drug for patients with uncomplicated hypertension. In addition to their antihypertensive effect, thiazides increase bone mineral density and reduce the prevalence of fractures. Traditionally, these effects have been attributed to increased renal calcium reabsorption that occurs secondary to the inhibition of the thiazide-sensitive sodium chloride cotransporter (NCC) in the distal tubule. The aim of the current study was to determine whether thiazides exert a direct bone-forming effect independent of their renal action. We found that the osteoblasts of human and rat bone also express NCC, suggesting that these bone-forming cells may be an additional target for thiazides. In vitro, NCC protein was virtually absent in proliferating human and fetal rat osteoblasts, whereas its expression dramatically increased during differentiation. Thiazides did not affect osteoblast proliferation, but directly stimulated the production of the osteoblast differentiation markers runt-related transcription factor 2 (runx2) and osteopontin. Using overexpression/knockdown studies in fetal rat calvarial cells, we show that thiazides increase the formation of mineralized nodules, but loop diuretics do not. Overall, our study demonstrates that thiazides directly stimulate osteoblast differentiation and bone mineral formation independent of their effects in the kidney. Therefore, in addition to their use as antihypertensive drugs, our results suggest that thiazides may find a role in the prevention and treatment of osteoporosis.


Subject(s)
Bone and Bones/metabolism , Calcification, Physiologic , Osteoblasts/pathology , Osteogenesis/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology , Sodium Chloride Symporters/metabolism , Animals , Biomarkers , Cell Differentiation/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mice , Osteopontin/metabolism , Rats , Rats, Sprague-Dawley , Sodium Chloride Symporters/drug effects
3.
Endocrinology ; 148(7): 3156-63, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17412806

ABSTRACT

Changes in extracellular [Ca2+] modulate the function of bone cells in vitro via the extracellular Ca2+-sensing receptor (CaR). Within bone microenvironments, resorption increases extracellular [Ca2+] locally. To determine whether enhanced CaR signaling could modulate remodeling and thereby bone mass in vivo, we generated transgenic mice with a constitutively active mutant CaR (Act-CaR) targeted to their mature osteoblasts by the 3.5 kb osteocalcin promoter. Longitudinal microcomputed tomography of cancellous bone revealed reduced bone volume and density, accompanied by a diminished trabecular network, in the Act-CaR mice. The bone loss was secondary to an increased number and activity of osteoclasts, demonstrated by histomorphometry of secondary spongiosa. Histomorphometry, conversely, indicates that bone formation rates were unchanged in the transgenic mice. Constitutive signaling of the CaR in mature osteoblasts resulted in increased expression of RANK-L (receptor activator of nuclear factor-kappaB ligand), the major stimulator of osteoclast differentiation and activation, which is the likely underlying mechanism for the bone loss. The phenotype of Act-CaR mice is not attributable to systemic changes in serum [Ca2+] or PTH levels. We provide the first in vivo evidence that increased signaling by the CaR in mature osteoblasts can enhance bone resorption and further propose that fluctuations in the [Ca2+] within the bone microenvironment may modulate remodeling via the CaR.


Subject(s)
Bone and Bones/metabolism , Osteoblasts/metabolism , Receptors, Calcium-Sensing/physiology , Animals , Bone Density/genetics , Bone Resorption/blood , Bone Resorption/genetics , Bone Resorption/physiopathology , Bone and Bones/pathology , Calcium/blood , Cattle , Cells, Cultured , Female , Immunoblotting , Inositol Phosphates/metabolism , Male , Mice , Mice, Transgenic , Mutation , Osteoblasts/cytology , Parathyroid Hormone/blood , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/physiology , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tomography, X-Ray Computed
4.
J Biol Chem ; 282(20): 15048-56, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17376781

ABSTRACT

The agonist sensitivity of the calcium-sensing receptor (CaR) can be altered by protein kinase C (PKC), with CaR residue Thr(888) contributing significantly to this effect. To determine whether CaR(T888) is a substrate for PKC and whether receptor activation modulates such phosphorylation, a phospho-specific antibody against this residue was raised (CaR(pT888)). In HEK-293 cells stably expressing CaR (CaR-HEK), but not in cells expressing the mutant receptor CaR(T888A), phorbol ester (PMA) treatment increased CaR(pT888) immunoreactivity as observed by immunoblotting and immunofluorescence. Raising extracellular Ca(2+) concentration from 0.5 to 2.5 mM increased CaR(T888) phosphorylation, an effect that was potentiated stereoselectively by the calcimimetic NPS R-467. These responses were mimicked by 5 mM extracellular Ca(2+) and abolished by the calcilytic NPS-89636 and also by PKC inhibition or chronic PMA pretreatment. Whereas CaR(T888A) did exhibit increased apparent agonist sensitivity, by converting intracellular Ca(2+) (Ca(2+)(i)) oscillations to sustained plateau responses in some cells, we still observed Ca(2+)(i) oscillations in a significant number of cells. This suggests that CaR(T888) contributes significantly to CaR regulation but is not the exclusive determinant of CaR-induced Ca(2+)(i) oscillations. Finally, dephosphorylation of CaR(T888) was blocked by the protein phosphatase 1/2A inhibitor calyculin, a treatment that also inhibited Ca(2+)(i) oscillations. In addition, calyculin/PMA cotreatment increased CaR(T888) phosphorylation in bovine parathyroid cells. Therefore, CaR(T888) is a substrate for receptor-induced, PKC-mediated feedback phosphorylation and can be dephosphorylated by a calyculin-sensitive phosphatase.


Subject(s)
Aniline Compounds/pharmacology , Calcium Signaling/drug effects , Enzyme Inhibitors/pharmacology , Parathyroid Glands/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Receptors, Calcium-Sensing/metabolism , Animals , Calcium/metabolism , Calcium/pharmacology , Calcium Signaling/genetics , Carcinogens/pharmacology , Cattle , Cell Line , Gene Expression , Humans , Mutation , Parathyroid Glands/cytology , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Protein Phosphatase 1 , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Receptors, Calcium-Sensing/genetics , Tetradecanoylphorbol Acetate/pharmacology
6.
Cell Calcium ; 35(3): 249-55, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15200148

ABSTRACT

Bone is the major sink and store for calcium and it fulfils essential roles in the maintenance of extracellular free ionised calcium concentration ([Ca2+]e) within its homeostatic range (1.1-1.3 mM). In response to acute hypercalcaemia or hypocalcaemia, Ca2+ is rapidly transported into or out of bone. Bone turnover (and therefore bone Ca2+ turnover) achieves the long-term correction of the [Ca2+]e by the metabolic actions of osteoblasts and osteoclasts, as they respectively incorporate or release Ca2+ from bone. These processes are regulated by the actions of hormones, such as parathyroid hormone (PTH), the release of which is a function of the [Ca2+]e, and is regulated by the action of the Ca2+-sensing receptor (CaR) in the parathyroid gland. Tissue culture studies indicate that bone cells also directly respond to increasing and decreasing [Ca2+]e in their vicinity, independently of the systemic factors. Nevertheless, further studies are necessary to identify how the acute and long-term local changes in [Ca2+]e affect bone cells and the physiological processes they are involved in. Also, the molecular mechanisms which enable the bone cells to sense and respond to [Ca2+]e are not clear. Like the parathyroid cells, bone cells also express the CaR, and accumulating evidence indicates the involvement of this receptor in their responses to the changing extracellular ionic environment.


Subject(s)
Bone and Bones/physiology , Calcium/metabolism , Receptors, Calcium-Sensing/physiology , Animals , Bone Remodeling/physiology , Bone Resorption/metabolism , Extracellular Space/metabolism , Homeostasis/physiology , Humans , Hyperparathyroidism/drug therapy , Ion Channels/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Osteocytes/physiology , Osteoporosis/drug therapy , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 101(14): 5140-5, 2004 Apr 06.
Article in English | MEDLINE | ID: mdl-15051872

ABSTRACT

We investigated the direct effects of changes in free ionized extracellular calcium concentrations ([Ca2+]o) on osteoblast function and the involvement of the calcium-sensing receptor (CaR) in mediating these responses. CaR mRNA and protein were detected in osteoblast models, freshly isolated fetal rat calvarial cells and murine clonal osteoblastic 2T3 cells, and in freshly frozen, undecalcified preparations of human mandible and rat femur. In fetal rat calvarial cells, elevating [Ca2+]o and treatment with gadolinium, a nonpermeant CaR agonist, resulted in phosphorylation of the extracellular signal-regulated kinases 1 and 2, Akt, and glycogensynthase kinase 3beta, consistent with signals of cell survival and proliferation. In agreement, cell number was increased under these conditions. Expression of the osteoblast differentiation markers core binding factor alpha1, osteocalcin, osteopontin, and collagen I mRNAs was increased by high [Ca2+]o, as was mineralized nodule formation. Alkaline phosphatase activity was maximal for [Ca2+]o between 1.2 and 1.8 mM. Inhibition of CaR by NPS 89636 blocked responses to the CaR agonists. In conclusion, we show that small deviations of [Ca2+]o from physiological values have a profound impact on bone cell fate, by means of the CaR and independently of systemic calciotropic peptides.


Subject(s)
Calcium/metabolism , Osteoblasts/physiology , Animals , Base Sequence , Bone and Bones/metabolism , Cell Division , DNA Primers , Extracellular Space/metabolism , Gene Expression , Humans , Immunohistochemistry , Osteoblasts/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...