Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 74(3): 438-47, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17531955

ABSTRACT

Strontium ranelate has several beneficial effects on bone and reduces the risk of vertebral and hip fractures in women with postmenopausal osteoporosis. We investigated whether Sr(2+) acts via a cell surface calcium-sensing receptor (CaR) in HEK293 cells stably transfected with the bovine CaR (HEK-CaR) and rat primary osteoblasts (POBs) expressing the CaR endogenously. Elevating Ca(o)(2+) or Sr(2+) concentration-dependently activated the CaR in HEK-CaR but not in non-transfected cells, but the potency of Sr(2+) varied depending on the biological response tested. Sr(2+) was less potent than Ca(o)(2+) in stimulating inositol phosphate accumulation and in increasing Ca(i)(2+), but was comparable to Ca(o)(2+) in stimulating ERK phosphorylation and a non-selective cation channel, suggesting that Ca(2+) and Sr(2+) have differential effects on specific cellular processes. With physiological concentrations of Ca(o)(2+), Sr(2+)-induced further CaR activation. Neither Sr(2+) nor Ca(o)(2+) affected the four parameters just described in non-transfected cells. In POB, Sr(2+) stimulated cellular proliferation. This effect was CaR-mediated, as transfecting the cells with a dominant negative bovine CaR significantly attenuated Ca(o)(2+)-stimulated POB proliferation. Finally, Sr(2+) significantly increased the mRNA levels of the immediate early genes, c-fos and egr-1, which are involved in POB proliferation, and this effect was attenuated by overexpressing the dominant negative CaR. In conclusion, Sr(2+) is a full CaR agonist in HEK-CaR and POB, and, therefore, the anabolic effect of Sr(2+) on bone in vivo could be mediated, in part, by the CaR.


Subject(s)
Cell Proliferation/drug effects , Organometallic Compounds/pharmacology , Osteoblasts/drug effects , Receptors, Calcium-Sensing/physiology , Thiophenes/pharmacology , Animals , Base Sequence , Calcium/pharmacology , Cell Line , DNA Primers , Humans , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/cytology , Patch-Clamp Techniques , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley
2.
Biochem Biophys Res Commun ; 354(1): 8-13, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17223073

ABSTRACT

The calcium-sensing receptor (CaR) mediates the effects of extracellular calcium ([Ca(2+)](o)) on PTH release, such that increasing levels of [Ca(2+)](o) inhibit PTH secretion through poorly defined mechanisms. In the present studies, immunocytochemical analysis demonstrated that F-actin, PTH, CaR, and caveolin-1 are colocalized at the apical secretory pole of PT cells, and subcellular fractionation of PT cells showed these proteins to be present within the secretory granule fraction. High [Ca(2+)](o) caused F-actin, PTH, and caveolin-1 to move to the apical pole of the cells. Depolymerization of F-actin by cytochalasin reduced the actin network and induced redistribution of actin/caveolin-1 to a dispersed pattern within the cell. The F-actin-severing compounds, latrunculin and cytochalasin, significantly increased PTH secretion, while the actin polymerizing agent, jasplakinolide, substantially inhibited PTH secretion. We have demonstrated that in polarized PT cells, the F-actin cytoskeleton is involved in the regulation of PTH secretion and is critical for inhibition of PTH secretion by high calcium.


Subject(s)
Calcium/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Parathyroid Glands/metabolism , Parathyroid Hormone/metabolism , Secretory Vesicles/metabolism , Animals , Cattle , Cells, Cultured , Extracellular Fluid/metabolism
3.
Mol Endocrinol ; 19(10): 2603-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15928311

ABSTRACT

The properties of neoplastic proliferation and hormonal dysregulation are tightly linked in primary hyperparathyroidism (HPT). However, whether abnormal parathyroid proliferation is the cause or result of a shift in calcium-sensitive parathyroid hormonal regulation has been controversial. We addressed this issue by analyzing the temporal sequence of these fundamental abnormalities in a mouse model of primary HPT. These transgenic mice (PTH-D1) harbor a transgene that targets overexpression of the cyclin D1 oncogene to parathyroid cells, resulting in parathyroid hypercellularity with a phenotype of chronic biochemical HPT and, notably, an abnormal in vivo PTH-calcium set point. We examined parathyroid cell proliferation and biochemical alterations in PTH-D1 and control wild-type mice from ages 1-14 months. Strikingly, abnormal parathyroid proliferation regularly preceded dysregulation of the calcium-PTH axis, supporting the concept that disturbed parathyroid proliferation is the crucial primary initiator leading to the development of the biochemical phenotype of HPT. Furthermore, we observed that decreased expression of the calcium-sensing receptor in the parathyroid glands occurs several months before development of biochemical HPT, suggesting that decreased calcium-sensing receptor may not be sufficient to cause PTH dysregulation in this animal model of primary HPT.


Subject(s)
Hyperparathyroidism/metabolism , Hyperparathyroidism/pathology , Animals , Calcium/metabolism , Cell Proliferation , Disease Models, Animal , Gene Expression , Genes, bcl-1 , Hyperparathyroidism/etiology , Mice , Mice, Transgenic , Parathyroid Hormone/metabolism , Phenotype , Receptors, Calcium-Sensing/metabolism , Time Factors
4.
Bone ; 35(3): 664-72, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15336602

ABSTRACT

We have previously reported that high extracellular Ca2+ stimulates parathyroid hormone-related protein (PTHrP) release from human prostate and breast cancer cell lines as well as from H-500 rat Leydig cancer cells, an action mediated by the calcium-sensing receptor (CaR). Activating the CaR leads to phosphorylation of mitogen-activated protein kinases (MAPKs) that participate in PTHrP synthesis and secretion. Because the CaR is a G protein-coupled receptor (GPCR), it is likely to transactivate the epidermal growth factor receptor (EGFR) or the platelet-derived growth factor receptor (PDGFR). In this study, we hypothesized that activation of the CaR transactivates the EGFR or PDGFR, and examined whether transactivation affects PTHrP secretion in PC-3 human prostate cancer cells. Using Western analysis, we observed that an increase in extracellular Ca2+ resulted in delayed activation of extracellular signal-regulated kinase (ERK) in PC-3 cells. Pre-incubation with AG1478 (an EGFR kinase inhibitor) or an EGFR neutralizing antibody inhibited the high Ca2+ -induced phosphorylation of ERK1/2. GM6001, a pan matrix metalloproteinase (MMP) inhibitor, also partially suppressed the ERK activation, but AG1296 (a PDGFR kinase inhibitor) did not. High extracellular Ca2+ stimulates PTHrP release during a 6-h incubation (1.5- to 2.5- and 3- to 4-fold increases in 3.0 and 7.5 mM Ca2+, respectively). When cells were preincubated with AG1478, GM6001, or an antihuman heparin-binding EGF (HB-EGF) antibody, PTHrP secretion was significantly inhibited under basal as well as high Ca2+ conditions, while AG1296 had no effect on PTHrP secretion. Taken together, these findings indicate that activation of the CaR transactivates the EGFR, but not the PDGFR, leading to phosphorylation of ERK1/2 and resultant PTHrP secretion, although CaR-EGFR-ERK might not be the only signaling pathway for PTHrP secretion. This transactivation is most likely mediated by activation of MMP and cleavage of proheparin-binding EGF (proHB-EGF) to HB-EGF.


Subject(s)
ErbB Receptors/physiology , Parathyroid Hormone-Related Protein/metabolism , Prostatic Neoplasms/metabolism , Receptors, Calcium-Sensing/metabolism , Transcriptional Activation/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Parathyroid Hormone-Related Protein/genetics , Prostatic Neoplasms/genetics , Receptors, Calcium-Sensing/genetics
6.
J Clin Endocrinol Metab ; 89(2): 548-56, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14764760

ABSTRACT

Autoimmune hypoparathyroidism is thought to result from immune-mediated destruction of the parathyroid glands. We encountered two patients with hypoparathyroidism and other autoimmune conditions (Graves' disease and Addison's disease, respectively) in whom autoimmune destruction of the parathyroid glands had not taken place. In the first, a histologically normal parathyroid gland was observed at the time of subtotal thyroidectomy; and in the second, the hypoparathyroidism remitted spontaneously. Both patients had antibodies that reacted with the cell surface of bovine parathyroid cells and human embryonic kidney (HEK293) cells transfected with the extracellular calcium-sensing receptor (CaR) but not with nontransfected HEK293 cells. The antibodies also reacted with the same bands on Western analysis of extracts of bovine parathyroid tissue and CaR-transfected HEK293 cells that were identified by an authentic, polyclonal, anti-CaR antiserum and reacted with several peptides with sequences from the CaR's extracellular domain. These anti-CaR antibodies activated the receptor based on their ability to increase inositol phosphate accumulation, activate MAPK, and inhibit PTH secretion. These results, therefore, demonstrate that patients with the biochemical findings of primary hypoparathyroidism can harbor activating antibodies to the CaR, which, in the two cases studied here, did not produce irreversible destruction of the parathyroid glands.


Subject(s)
Autoantibodies/analysis , Autoantibodies/immunology , Autoimmune Diseases/immunology , Hypoparathyroidism/immunology , Receptors, Calcium-Sensing/immunology , Addison Disease/complications , Adult , Animals , Autoimmune Diseases/blood , Autoimmune Diseases/complications , Blotting, Western , Cattle , Cell Line , Graves Disease/complications , Humans , Hypoparathyroidism/blood , Hypoparathyroidism/complications , Inositol Phosphates/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Parathyroid Hormone/antagonists & inhibitors , Peptide Fragments/immunology , Precipitin Tests , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Transfection
7.
Eur J Endocrinol ; 150(1): 9-18, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14713274

ABSTRACT

OBJECTIVE: The pathogenesis of sporadic idiopathic hypoparathyroidism is unclear. The calcium sensing receptor (CaSR) plays a pivotal role in extracellular calcium homeostasis and is the candidate autoantigen in hypoparathyroidism associated with autoimmune polyglandular endocrinopathy syndrome. We therefore looked for antibodies (Ab) against the CaSR in patients with sporadic idiopathic hypoparathyroidism and their association, if any, with the major histocompatibility complex (MHC) class II human leukocyte antigen (HLA)-DR haplotypes. METHODS: The subjects included 51 patients with sporadic idiopathic hypoparathyroidism and 45 healthy controls. Investigations included computerised tomography, serum calcium, phosphorus, thyroxine, TSH, cortisol, intact parathyroid hormone (iPTH), ACTH and thyroid peroxidase (TPO) and adrenal antibodies. The CaSRAb were assayed in patients' sera by Western blot. Genotyping of the HLA-DR locus was performed using PCR and sequence-specific oligonucleotide probes. RESULTS: Intracranial calcification and cataract were present in 76.5% and 41.1% of the patients respectively and 62.7% had convulsions. Autoantibodies against the 168 kDa CaSR protein were demonstrated in the serum of 49.0% of the patients and in 13.3% of the controls (P<0.001). Pre-incubating serum samples from the CaSRAb-positive patients with parathyroid membrane produced a 90% decrease in the band intensity. HLA-DRB1*01 and DRB1*09 alleles were significantly associated with idiopathic hypoparathyroidism (relative risk of 7.8, P=0.001). The frequency of HLA-DRB1*09 and DRB1*10 alleles tended to be higher in patients positive for the CaSRAb. There was no significant difference in the frequency of occurrence of convulsions, cataract, intracranial calcification, calcium:phosphorus ratio, and iPTH levels between patients with and without CaSRAb. CONCLUSION: 49.0% of the patients studied had serological evidence of organ-specific autoimmunity against the CaSR protein. The occurrence of CaSRAb and the HLA-DR associations imply an autoimmune component to the disease, but the primary role of the CaSRAb in the pathogenesis of the disease needs to be assessed further.


Subject(s)
Autoantibodies/blood , Hypoparathyroidism/epidemiology , Hypoparathyroidism/immunology , Receptors, Calcium-Sensing/immunology , Adult , Aged , Animals , Female , HLA-DR Antigens/genetics , Haplotypes , Humans , Liver/immunology , Male , Myocardium/immunology , Rats , Seroepidemiologic Studies
8.
J Clin Endocrinol Metab ; 88(9): 4455-64, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970324

ABSTRACT

Caveolins are key components of caveolae membranes. The calcium-sensing receptor (CaR) resides within caveolin-rich membrane domains in bovine parathyroid (PT) cells. Recent studies reported reduced CaR expression, and abnormal calcium-sensing in PT tumors. To examine this altered CaR signaling, we investigated ERK activation after CaR stimulation in human and bovine PT cells. In freshly prepared bovine PT cells, high extracellular calcium (Ca(2+)(0)) stimulates ERK1/2 phosphorylation, and activated ERK1/2 colocalizes with caveolin-1 at the plasma membrane but fails to translocate to the nucleus, and cell proliferation is low. In cultured bovine PT cells, CaR and caveolin-1 levels are reduced; activated ERK1/2 localizes in the cell periphery at 10 min and in the perinuclear and nuclear regions at 60 min after exposure to high Ca(2+)(0), and cell proliferation is increased. In PT cells from adenomas, there are high levels of caveolin-2, variably reduced caveolin-1, and hyperactivation of ERK1/2, which colocalizes with caveolin-1 in some cells, but localizes in the cytosol and nucleus in others. Finally, caveolin-1 negative human PT cells exhibit reduced suppressibility of PTH secretion by high Ca(2+)(0). Thus, CaR and caveolin-1 colocalize in PT cells, and reduced levels of caveolin-1 could participate in the abnormal cellular function and proliferation of cultured bovine PT cells and PT adenomas.


Subject(s)
Adenoma/metabolism , Caveolins/biosynthesis , Mitogen-Activated Protein Kinases/biosynthesis , Parathyroid Glands/metabolism , Parathyroid Neoplasms/metabolism , Animals , Blotting, Western , Bromodeoxyuridine , Calcium Signaling/physiology , Cattle , Caveolin 1 , Caveolins/genetics , Cell Division/physiology , Cell Membrane/chemistry , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Calcium-Sensing , Receptors, Cell Surface/metabolism , Signal Transduction/physiology
9.
J Biol Chem ; 278(33): 31167-76, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12783889

ABSTRACT

The calcium-sensing receptor (CaR) is a G protein-coupled, seven-transmembrane receptor and resides within caveolin-rich membrane domains in bovine parathyroid cells. The proenzyme of calpain 2 (m-calpain) is a heterodimeric calcium-dependent cysteine protease consisting of catalytic and regulatory subunits. The effects of calcium on the enzyme include activation, autolysis, and subunit dissociation. Here, we examine the potential role of caveolin-1 and caveolae in regulating the cellular distribution and function of m-calpain in parathyroid cells. We show that the inactive heterodimeric forms of m-calpain are concentrated in caveolin-rich membrane fractions prepared from parathyroid cells incubated with low extracellular calcium (Ca2+(o)). In contrast, in cells incubated with 3 mm Ca2+(o), which activates the CaR and increases intracellular calcium, there is a reduction in m-calpain in association with an increase in CaR protein and phosphorylated protein kinase C alpha and beta in caveolin-rich fractions. To assess the impact of activation of calpain on CaR protein in caveolar fractions, we analyzed the effects of m-calpain on the CaR. Activation of the CaR with high Ca2+(o) induced the release of lower molecular weight fragments of the receptor into the cell culture medium, and calpain inhibitors blocked this effect. Moreover, the fragments of the CaR as well as caveolin-1, m-calpain, and alkaline phosphatase were localized in membrane vesicles shed by parathyroid cells, supporting the association of these proteins in living cells. Treatment of CaR proteins in vitro with m-calpain also resulted in the appearance of lower molecular weight fragments of the CaR. Our data suggest that localization of m-calpain within caveolae may contribute to maintenance of the enzyme in an inactive state and that m-calpain may also contribute to the regulation of CaR levels.


Subject(s)
Calpain/metabolism , Parathyroid Glands/enzymology , Receptors, Cell Surface/metabolism , Animals , Autolysis , Calcium/pharmacology , Calpain/analysis , Carcinogens/pharmacology , Cattle , Caveolae/metabolism , Caveolin 1 , Caveolins/analysis , Cell Line , Humans , Isoenzymes/metabolism , Parathyroid Glands/chemistry , Parathyroid Glands/cytology , Parathyroid Hormone/analysis , Phosphorylation , Protein Kinase C/metabolism , Rabbits , Receptors, Calcium-Sensing , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology
10.
J Clin Endocrinol Metab ; 88(1): 60-72, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12519831

ABSTRACT

Antibodies to cell surface receptors can cause endocrine dysfunction by mimicking or blocking the actions of their respective hormones. We sought patients with autoantibodies to the extracellular calcium (Ca(2+)(o))-sensing receptor (CaR), which sets the normal level of blood calcium, that mimic the genetic disorder, familial hypocalciuric hypercalcemia, caused by heterozygous inactivating mutations of the CaR. Four individuals from two kindreds were identified with PTH-dependent hypercalcemia, who had other autoimmune manifestations: one with sprue and antigliadin and antiendomyseal antibodies and three with antithyroid antibodies. Three of the patients also had relative or absolute hypocalciuria. The patients' sera contained antibodies that reacted with the cell surface of bovine parathyroid cells in a manner similar to an authentic polyclonal anti-CaR antibody, stained bands on Western analysis of sizes similar to those labeled by the anti-CaR antiserum, and reacted with several synthetic peptides derived from sequences within the CaR's extracellular amino terminus. The patients' sera also stimulated PTH release from dispersed human parathyroid cells compared with the effect of sera from normocalcemic control subjects. This stimulation could be blocked by preabsorbing serum with membranes from CaR-transfected, but not nontransfected, human embryonic kidney (HEK293) cells. Finally, in two of the patients, antibodies affinity-purified using a synthetic peptide from within the CaR's extracellular domain inhibited high Ca(2+)(o)-stimulated, CaR-mediated accumulation of inositol phosphates and activation of mitogen-activated protein kinase in CaR-transfected HEK293 cells. DNA sequencing revealed no mutations within the index patients' CaR genes in the two families. Therefore, a biochemical phenotype of PTH-dependent hypercalcemia resembling that caused by heterozygous inactivating mutations of the CaR in familial hypocalciuric hypercalcemia can be observed in patients with antibodies to the CaR's extracellular domain that stimulate PTH release, probably by inhibiting activation of the CaR by Ca(2+)(o). Autoimmune hypocalciuric hypercalcemic is an acquired disorder of Ca(2+)(o) sensing that should be differentiated from that caused by inactivating mutations of the CaR.


Subject(s)
Autoantibodies/immunology , Calcium/urine , Hypercalcemia/immunology , Receptors, Cell Surface/immunology , Adult , Autoantibodies/blood , Autoantibodies/physiology , Blood Physiological Phenomena , Blotting, Western , Calcium/physiology , Cell Line , Enzyme Activation/physiology , Extracellular Space/metabolism , Female , Humans , Hypercalcemia/genetics , Hypercalcemia/physiopathology , Immunoglobulins/metabolism , Inositol Phosphates/metabolism , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Parathyroid Glands/immunology , Parathyroid Glands/pathology , Parathyroid Hormone/metabolism , Pedigree , Peptide Fragments/immunology , Receptors, Calcium-Sensing , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Syndrome
11.
J Biol Chem ; 277(52): 50543-9, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12409307

ABSTRACT

The extracellular calcium (Ca(2+)(o))-sensing receptor (CaR) activates Ca(2+) influx independent of the release of intracellular Ca(2+) stores. The latter can be negatively regulated by protein kinase C (PKC) through phosphorylation of Thr-888 of the CaR. In this study, we substituted Thr-888 with various amino acid residues or a stop codon to understand how PKC phosphorylation of the CaR inhibits receptor-mediated release of intracellular Ca(2+) stores. Substitutions of Thr-888 with hydrophobic and hydrophilic amino acid residues had various effects on CaR-mediated release of intracellular Ca(2+) stores as well as activation of Ca(2+) influx. Several point mutations, such as T888D, had marked negative effects on CaR-mediated release of intracellular Ca(2+) stores but not on phorbol myristate acetate-insensitive activation of Ca(2+) influx. Presumably, the negatively charged aspartate mimics phospho-threonine. Interestingly, truncating the receptor at 888 had an even more pronounced negative effect on CaR-elicited release of intracellular Ca(2+) stores without significantly affecting CaR-mediated activation of Ca(2+) influx. Therefore, truncation at position 888 of the CaR affects the activity of the receptor in a manner that resembles PKC phosphorylation of the CaR. This in turn suggests that PKC phosphorylation of the CaR prevents G protein subtypes from interacting with the region of the receptor critical for releasing Ca(2+) stores, which is missing in the truncated receptor.


Subject(s)
Calcium Signaling/physiology , GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Substitution , Binding Sites , Calcium/pharmacology , Cell Line , Humans , Kinetics , Mutagenesis, Site-Directed , Phosphorylation , Point Mutation , Receptors, Calcium-Sensing , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Spermine/pharmacology , Threonine , Transfection
12.
Curr Opin Nephrol Hypertens ; 11(4): 397-402, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12105389

ABSTRACT

PURPOSE OF REVIEW: To review recent developments regarding the mechanisms underlying the regulation of various aspects of parathyroid function. RECENT FINDINGS: New studies published during the past year focused on calcium sensing via the calcium-sensing receptor, signal transduction within parathyroid cells, regulation of parathyroid hormone secretion, and the role of caveolae in calcium-sensing receptor-mediated signal transduction. In recent years, in-vitro and in-vivo studies have suggested a dominant role for the calcium-sensing receptor in the regulation of not only parathyroid hormone secretion but also parathyroid cellular proliferation by extracellular calcium. The development of a mouse model for primary hyperparathyroidism that over expresses cyclin D1 in the parathyroid gland provides an experimental system for studying the molecular basis for reduced calcium receptor expression and its role in the pathophysiology of primary hyperparathyroidism. There is also increasing evidence for the importance of vitamin D and the level of inorganic phosphate in regulating parathyroid function. SUMMARY: Important advances are being made in understanding extracellular calcium- and calcium-sensing receptor-regulated signal transduction in the parathyroid but the subsequent steps coupling the calcium-sensing receptor to the control of parathyroid hormone secretion and parathyroid cellular proliferation remain to be fully elucidated.


Subject(s)
Parathyroid Glands/physiology , Signal Transduction/physiology , Animals , Caveolae/physiology , Humans , Parathyroid Hormone/metabolism , Receptors, Calcium-Sensing , Receptors, Cell Surface/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...