Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
J Membr Biol ; 184(2): 113-20, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719848

ABSTRACT

The human epithelial channels, CaT1 and CaT2, were expressed in oocytes, and their single-channel characteristics were compared. In the presence of Na+ and K+ as charge carriers in the pipette solutions, channel activities were observed only when the the extracellular sides of the patches were exposed to nominally Ca2+- and Mg2+-free solutions. In patches of both CaT1- and CaT2-expressing oocytes, multiple channel openings were observed, but the current levels were higher in CaT2-expressing oocytes, particularly at more negative voltages. With K+ as a charge carrier in patches of CaT1-expressing oocytes, the channel activity was low at -10 to -60 mV, but increased dramatically at more negative potentials. This voltage dependence was observed in the presence of both Na+ and K+. The channel activity with Na+, however, was higher at all potentials. Differences between the voltage dependencies for the two cations were also observed in CaT2-expressing oocytes, but the channel activities were higher than those in CaT1-expressing oocytes, particularly in the presence of Na+. We also found that low concentrations of extracellular Mg2+ (5-50 microm) elicited a strong inhibitory action on the CaT channels. Activation of the CaT1 and CaT2 channels by hyperpolarization and other factors may promote increased Ca2+ entry that participates in stimulation of intestinal absorption and renal reabsorption and/or other Ca2+ transport mechanisms in epithelial cells.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Amino Acid Transport Systems, Basic , Animals , Calcium Channels/drug effects , Calcium Channels/genetics , Epithelial Cells/physiology , Humans , Magnesium/pharmacology , Membrane Potentials/physiology , Oocytes/metabolism , Potassium/metabolism , RNA, Complementary/genetics , Rats , Sodium/metabolism , TRPV Cation Channels , Transfection , Xenopus laevis
2.
Biochem Biophys Res Commun ; 282(5): 1251-6, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11302751

ABSTRACT

Most patients with autosomal dominant polycystic kidney disease (ADPKD) harbor mutations truncating polycystin-1 (PC1) or polycystin-2 (PC2), products of the PKD1 and PKD2 genes, respectively. A third member of the polycystin family, polycystin-L (PCL), was recently shown to function as a Ca(2+)-modulated nonselective cation channel. More recently, PC2 was also shown to be a nonselective cation channel with comparable properties to PCL, though the membrane targeting of PC2 likely varies with cell types. Here we show that PC2 expressed heterologously in Xenopus oocytes is targeted to intracellular compartments. By contrast, a truncated form of mouse PC2 corresponding to a naturally occurring human mutation R742X is targeted predominantly to the plasma membrane where it mediates K(+), Na(+), and Ca(2+) currents. Unlike PCL, the truncated form does not display Ca(2+)-activated transport activities, possibly due to loss of an EF-hand at the C-terminus. We propose that PC2 forms ion channels utilizing structural components which are preserved in the R742X form of the protein. Implications for epithelial cell signaling are discussed.


Subject(s)
Amino Acid Substitution , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation/genetics , Amino Acid Motifs/physiology , Animals , Calcium/metabolism , Calcium/pharmacokinetics , Cell Compartmentation/physiology , Cell Membrane/metabolism , Cells, Cultured , Intracellular Fluid/metabolism , Ion Channels/physiology , Membrane Potentials/physiology , Mice , Microinjections , Patch-Clamp Techniques , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Protein Transport/physiology , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , TRPP Cation Channels , Xenopus
3.
Biochem Biophys Res Commun ; 282(1): 341-50, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11264013

ABSTRACT

Mutations in polycystins-1 and -2 (PC1 and PC2) cause autosomal dominant polycystic kidney disease (ADPKD), which is characterized by progressive development of epithelial renal cysts, ultimately leading to renal failure. The functions of these polycystins remain elusive. Here we show that PC2 is a Ca(2+)-permeable cation channel with properties distinct from any known intracellular channels. Its kinetic behavior is characterized by frequent transitions between closed and open states over a wide voltage range. The activity of the PC2 channel is transiently increased by elevating cytosolic Ca(2+). Given the predominant endoplasmic reticulum (ER) location of PC2 and its unresponsiveness to the known modulators of mediating Ca(2+) release from the ER, inositol-trisphosphate (IP(3)) and ryanodine, these results suggest that PC2 represents a novel type of channel with properties distinct from those of the other Ca(2+)-release channels. Our data also show that the PC2 channel can be translocated to the plasma membranes by defined chemical chaperones and proteasome modulators, suggesting that in vivo, it may also function in the plasma membrane under specific conditions. The sensitivity of the PC2 channel to changes of intracellular Ca(2+) concentration is deficient in a mutant found in ADPKD patients. The dysfunction of such mutants may result in defective coupling of PC2 to intracellular Ca(2+) homeostasis associated with the pathogenesis of ADPKD.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Homeostasis/physiology , Membrane Proteins/physiology , Polycystic Kidney Diseases/physiopathology , Animals , Calcium Channels/genetics , Cloning, Molecular , DNA, Complementary , Humans , Immunohistochemistry , Membrane Proteins/genetics , Mice , TRPP Cation Channels , Xenopus
4.
Am J Physiol Cell Physiol ; 280(2): C382-93, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208534

ABSTRACT

We have previously shown the expression of the extracellular calcium (Ca2+o)-sensing receptor (CaR) in osteoblast-like cell lines, and others have documented its expression in sections of murine, bovine, and rat bone. The existence of the CaR in osteoblasts remains controversial, however, since some studies have failed to document its expression in the same osteoblast-like cell lines. The goals of the present study were twofold. 1) We sought to determine whether the CaR is expressed in the human osteoblast-like cell line, MG-63, which has recently been reported by others not to express this receptor. 2) We investigated whether the CaR, if present in MG-63 cells, is functionally active, since most previous studies have not proven the role of the CaR in mediating known actions of Ca2+o on osteoblast-like cells. We used immunocytochemistry and Western blotting with the specific, affinity-purified anti-CaR antiserum 4637 as well as Northern blot analysis and RT-PCR using a riboprobe and PCR primers specific for the human CaR, respectively, to show readily detectable CaR protein and mRNA expression in MG-63 cells. Finally, we employed the patch-clamp technique to show that an elevation in Ca2+o as well as the specific, allosteric CaR activator NPS R-467 (0.5 microM), but not its less active stereoisomer NPS S-467 (0.5 microM), activate an outward K+ channel in MG-63 cells, strongly suggesting that the CaR in MG-63 cells is not only expressed but is functionally active.


Subject(s)
Calcium/metabolism , Osteoblasts/metabolism , Receptors, Cell Surface/metabolism , Aniline Compounds/pharmacology , Cell Membrane/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Potassium Channels/drug effects , Potassium Channels/physiology , Receptors, Calcium-Sensing , Tumor Cells, Cultured
5.
Biochem Biophys Res Commun ; 280(1): 145-50, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11162491

ABSTRACT

A newly cloned, human epithelial Ca2+ transport protein (CaT1) was expressed in Xenopus laevis oocytes, and its single channel characteristics were examined. The CaT1 channel shows a strong dependence upon hyperpolarizing voltages, being activated by very negative voltages. The probability of channel opening and mean open times increase substantially at more negative voltages in the range of -90 to -160 mV. In addition, CaT1 channel activity was markedly inhibited by micromolar levels of a noncompetitive antagonist of the IP3 receptor originally isolated from a marine sponge, Xestospongin C. This inhibitory effect could be mediated indirectly via the binding of Xestospongin C to the inositol-trisphosphate (IP3) receptor or, alternatively, by a direct action on the CaT1 channel itself. Independent of its mechanism of action in inhibiting CaT1, Xestospongin C will provide a useful tool for elucidating the physiological role(s) of this novel epithelial Ca2+ channel.


Subject(s)
Calcium Channels/physiology , Chelating Agents/pharmacology , Edetic Acid/analogs & derivatives , Oxazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Animals , Calcium Channels/drug effects , Edetic Acid/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Inositol 1,4,5-Trisphosphate Receptors , Macrocyclic Compounds , Membrane Potentials/drug effects , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , TRPV Cation Channels , Xenopus laevis
6.
Biochem Biophys Res Commun ; 278(2): 326-32, 2000 Nov 19.
Article in English | MEDLINE | ID: mdl-11097838

ABSTRACT

Transcellular calcium transport occurs in many epithelial tissues including intestine, kidney, and placenta. We identified the human ortholog (hCaT1) of a recently cloned rat calcium transport protein, CaT1, that mediates intestinal calcium uptake. hCaT1 messenger RNA is present in the gastrointestinal tract, including esophagus, stomach, duodenum, jejunum, ileum, and colon. High levels of hCaT1 transcripts are also present in pancreas, placenta, prostate, and salivary gland, while moderate levels are present in liver, kidney, and testis. hCaT1 mRNA is also expressed in the colorectal cancer cell line, SW480, and the chronic myelogenous leukemia cell line, K-562. The hCaT1 gene was assigned to the long arm of chromosome 7, bands q33-34, by fluorescence in situ hybridization. When expressed in Xenopus laevis oocytes, hCaT1 promotes saturable Ca(2+) uptake with a Michaelis constant of 0.25 mM. Our studies suggest a role for hCaT1 in cellular calcium uptake in a variety of tissues, including the transcellular calcium transport pathway in intestine.


Subject(s)
Calcium Channels , Calcium/metabolism , Catalase/metabolism , Proteins , Amino Acid Sequence , Animals , Catalase/chemistry , Catalase/genetics , Catalase/physiology , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 7 , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , TRPV Cation Channels , Xenopus laevis
7.
Brain Res Mol Brain Res ; 80(2): 177-87, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11038250

ABSTRACT

Glioblastoma cells exhibit several forms of sensitivity to extracellular calcium (Ca(o)) that might be conferred by the Ca(o)-sensing receptor (CaR) that is intimately involved in the maintenance of Ca(o) homeostasis by various cell types. This receptor is expressed in human glioblastoma cell line, U87, and here we show that CaR activators stimulate a Ca(2+)-activated potassium (K(+)) channel (CAKC) with a conductance of 140 pS. The responses to CaR activators, however, were blunted in U87 cells transfected with a CaR bearing an inactivating mutation (R185Q) that has previously been shown to exert a dominant negative (DN) action on the wild type receptor. Raising Ca(o) from 0.75 to 2.0 mM or addition of a polycationic CaR agonist, each activated CAKC in nontransfected wild type and empty vector-transfected U87 cells, while they had little or no effect on channel activity in cells expressing the DN CaR (DN-CaR cells). In nontransfected wild type and empty vector-transfected cells, the specific 'calcimimetic' CaR activator, NPS R-467, stimulated the channel, while its less active stereoisomer, NPS S-467, did not. In DN-CaR cells, in contrast, NPS R-467, had no effect on channel activity, suggesting defective coupling of the CaR to this ion channel. CaR-mediated stimulation of these K(+) channels could lead to membrane repolarization and related changes in cellular function under normal conditions. Since the R185Q mutation in the CaR produces a more severe phenotype in humans than most inactivating mutations of this receptor, some of its clinical consequences could potentially result from abnormal CaR-dependent channel functioning.


Subject(s)
Calcium/pharmacology , Glioblastoma , Potassium Channels/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Calcium/metabolism , Extracellular Space/chemistry , Extracellular Space/metabolism , Gene Expression Regulation, Neoplastic , Homeostasis/physiology , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutagenesis/physiology , Patch-Clamp Techniques , Potassium/metabolism , Receptors, Calcium-Sensing , Transfection , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/metabolism
8.
J Biol Chem ; 275(36): 28186-94, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10875938

ABSTRACT

Active absorption of calcium from the intestine and reabsorption of calcium from the kidney are major determinants of whole body calcium homeostasis. Two recently cloned proteins, CaT1 and ECaC, have been postulated to mediate apical calcium uptake by rat intestine and rabbit kidney, respectively. By screening a rat kidney cortex library with a CaT1 probe, we isolated a cDNA encoding a protein (CaT2) with 84.2 and 73.4% amino acid identities to ECaC and CaT1, respectively. Unlike ECaC, CaT2 is kidney-specific in the rat and was not detected in intestine, brain, adrenal gland, heart, skeletal muscle, liver, lung, spleen, thymus, and testis by Northern analysis or reverse transcription polymerase chain reaction. The expression pattern of CaT2 in kidney was similar to that of calbindin D(28K) and the sodium calcium exchanger 1, NCX1, by in situ hybridization of adjacent sections. Furthermore, the mRNAs for CaT2 and calbindin D(28K) were colocalized in the same cells. CaT2 mediated saturable calcium uptake with a Michaelis constant (K(m)) of 0.66 mm when expressed in Xenopus laevis oocytes. Under voltage clamp condition, CaT2 promoted inward currents in X. laevis oocytes upon external application of Ca(2+). Sr(2+) and Ba(2+) but not Mg(2+) also evoked inward currents in CaT2-expressing oocytes. Similar to the alkaline earth metal ions, application of Cd(2+) elicited inward current in CaT2-expressing oocytes with a K(m) of 1.3 mm. Cd(2+), however, also potently inhibited CaT2-mediated Ca(2+) uptake with an IC(50) of 5.4 micrometer. Ca(2+) evoked currents were reduced at low pH and increased at high pH and were only slightly affected by the L-type voltage-dependent calcium channel antagonists, nifedipine, verapamil, diltiazem, and the agonist, Bay K 8644, even at relatively high concentrations. In conclusion, CaT2 may participate in calcium entry into the cells of the distal convoluted tubule and connecting segment of the nephron, where active reabsorption of calcium takes place via the transcellular route. The high sensitivity of CaT2 to Cd(2+) also provides a potential explanation for Cd(2+)-induced hypercalciuria and resultant renal stone formation.


Subject(s)
Calcium Channels/physiology , Kidney Cortex/metabolism , Nephrons/metabolism , Amino Acid Sequence , Amino Acid Transport Systems, Basic , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Calcium Channels/genetics , DNA, Complementary , Gene Library , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Molecular , Molecular Sequence Data , Oocytes/physiology , Protein Structure, Secondary , Rabbits , Rats , Sequence Alignment , Sequence Homology, Amino Acid , TRPV Cation Channels , Xenopus laevis
9.
Bone ; 27(1): 21-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10865205

ABSTRACT

The existence in osteoblasts of the G-protein-coupled extracellular calcium (Ca(o)(2+))-sensing receptor (CaR) that was originally cloned from parathyroid and kidney remains controversial. In our recent studies, we utilized multiple detection methods to demonstrate the expression of CaR transcripts and protein in several osteoblastic cell lines, including murine MC3T3-E1 cells. Although we and others have shown that high Ca(o)(2+) and other polycationic CaR agonists modulate the function of MC3T3-E1 cells, none of these actions has been unequivocally shown to be mediated by the CaR. Previous investigations using neurons and lens epithelial cells have shown that activation of the CaR stimulates Ca(2+)-activated K(+) channels. Because osteoblastic cells express a similar type of channel, we have examined the effects of specific "calcimimetic" CaR activators on the activity of a Ca(2+)-activated K(+) channel in MC3T3-E1 cells as a way of showing that the CaR is not only expressed in those cells but is functionally active. Patch-clamp analysis in the cell-attached mode showed that raising Ca(o)(2+) from 0.75 to 2.75 mmol/L elicited about a fourfold increase in the open state probability (P(o)) of an outward K(+) channel with a conductance of approximately 92 pS. The selective calcimimetic CaR activator, NPS R-467 (0.5 micromol/L), evoked a similar activation of the channel, while its less active stereoisomer, NPSS-467 (0.5 micromol/L), did not. Thus, the CaR is not only expressed in MC3T3-E1 cells, but is also functionally coupled to the activity of a Ca(2+)-activated K(+) channel. This receptor, therefore, could transduce local or systemic changes in Ca(o)(2+) into changes in the activity of this ion channel and related physiological processes in these and perhaps other osteoblastic cells.


Subject(s)
Osteoblasts/physiology , Potassium Channels/physiology , Receptors, Cell Surface/physiology , Animals , Cell Line , Ion Channel Gating/physiology , Mice , Patch-Clamp Techniques , Receptors, Calcium-Sensing , Signal Transduction
10.
Calcif Tissue Int ; 66(5): 375-82, 2000 May.
Article in English | MEDLINE | ID: mdl-10773108

ABSTRACT

Human promyelocytic leukemia cells (HL-60) have been used widely as a model for studying the differentiation of hematopoietic progenitor cells in vitro. After treatment with phorbol-12-myristate-13-acetate (PMA) or 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)], HL-60 cells differentiate into cells with the phenotype of monocytes/macrophages. We previously showed that peripheral blood monocytes and the murine J774 monocytic cell line express the CaR, and myeloid progenitors in the bone marrow and myeloid cells in peripheral blood other than monocytes express lower levels of the CaR. Therefore, we investigated whether undifferentiated HL-60 cells express a functional G protein-coupled, extracellular calcium (Ca(2+)(o))-sensing receptor (CaR) and if the expression of the CaR increases as these cells differentiate along the monocytic lineage. The use of reverse transcription-polymerase chain reaction (RT-PCR) with CaR-specific primers, followed by sequencing of the amplified products, identified an authentic CaR transcript in undifferentiated HL-60 cells. Both immunocytochemistry and Western blot analysis using a CaR-specific antiserum detected low levels of CaR protein expression in undifferentiated HL-60 cells. The levels of CaR protein increased considerably following treatment of the cells with PMA (50 nM) or 1,25(OH)(2)D(3) (100 nM) for 5 days. Northern analysis using a CaR-specific riboprobe identified CaR transcripts in undifferentiated HL-60 cells, but CaR mRNA levels did not change appreciably after treatment with either agent, suggesting that upregulation of CaR protein occurs at a translational level. PMA-treated HL-60 cells expressed a nonselective cation channel (NCC), and the calcimimetic CaR activator, NPS R-467, but not its less active stereoisomer, NPS S-467, as well as the polycationic CaR agonist, neomycin, activated this NCC, demonstrating that the CaR expressed in these cells is functionally active. Therefore, HL-60 cells exhibit an increase in CaR protein expression, occurring at a translational level during their differentiation into cells with a monocyte/macrophage phenotype in response to treatment with PMA or 1, 25(OH)(2)D(3), which is functionally linked to activation of a nonselective cation channel.


Subject(s)
Calcium/metabolism , Ion Channels/metabolism , Monocytes/metabolism , Receptors, Cell Surface/metabolism , Animals , Base Sequence , Cell Differentiation/drug effects , DNA Primers/genetics , HL-60 Cells , Humans , Immunohistochemistry , Mice , Monocytes/cytology , Monocytes/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcium-Sensing , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology
11.
Nature ; 401(6751): 383-6, 1999 Sep 23.
Article in English | MEDLINE | ID: mdl-10517637

ABSTRACT

Polycystic kidney diseases are genetic disorders in which the renal parenchyma is progressively replaced by fluid-filled cysts. Two members of the polycystin family (polycystin-1 and -2) are mutated in autosomal dominant polycystic kidney disease (ADPKD), and polycystin-L is deleted in mice with renal and retinal defects. Polycystins are membrane proteins that share significant sequence homology, especially polycystin-2 and -L (50% identity and 71% similarity). The functions of the polycystins remain unknown. Here we show that polycystin-L is a calcium-modulated nonselective cation channel that is permeable to sodium, potassium and calcium ions. Patch-clamp experiments revealed single-channel activity with a unitary conductance of 137 pS. Channel activity was substantially increased when either the extracellular or intracellular calcium-ion concentration was raised, indicating that polycystin-L may act as a transducer of calcium-mediated signalling in vivo. Its large single-channel conductance and regulation by calcium ions distinguish it from other structurally related cation channels.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Membrane Glycoproteins/metabolism , Phosphoproteins/metabolism , Animals , Calcium Channels/genetics , Calcium Signaling , Cations, Divalent/metabolism , Cell Membrane Permeability , Chelating Agents/pharmacology , Cloning, Molecular , Egtazic Acid/pharmacology , Electrophysiology , Humans , Membrane Glycoproteins/genetics , Patch-Clamp Techniques , Phosphoproteins/genetics , Polycystic Kidney Diseases/metabolism , Receptors, Cell Surface , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thapsigargin/pharmacology , Xenopus
12.
J Neurosci ; 19(20): 8876-84, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516307

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is thought to be caused in part by the age-related accumulation of amyloid beta-protein (Abeta). The presence of neuritic plaques containing abundant Abeta-derived amyloid fibrils in AD brain tissue supports the concept that fibril accumulation per se underlies neuronal dysfunction in AD. Recent observations have begun to challenge this assumption by suggesting that earlier Abeta assemblies formed during the process of fibrillogenesis may also play a role in AD pathogenesis. Here, we present the novel finding that protofibrils (PF), metastable intermediates in amyloid fibril formation, can alter the electrical activity of neurons and cause neuronal loss. Both low molecular weight Abeta (LMW Abeta) and PF reproducibly induced toxicity in mixed brain cultures in a time- and concentration-dependent manner. No increase in fibril formation during the course of the experiments was observed by either Congo red binding or electron microscopy, suggesting that the neurotoxicity of LMW Abeta and PF cannot be explained by conversion to fibrils. Importantly, protofibrils, but not LMW Abeta, produced a rapid increase in EPSPs, action potentials, and membrane depolarizations. These data suggest that PF have inherent biological activity similar to that of mature fibrils. Our results raise the possibility that the preclinical and early clinical progression of AD is driven in part by the accumulation of specific Abeta assembly intermediates formed during the process of fibrillogenesis.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Neurons/drug effects , Neurons/physiology , Neurotoxins/pharmacology , Amyloid beta-Peptides/analysis , Animals , Cerebral Cortex/pathology , Coloring Agents , Congo Red , Culture Media/chemistry , Electrophysiology , Microscopy, Electron , Molecular Weight , Protein Conformation , Rats , Time Factors
13.
J Mol Cell Cardiol ; 31(9): 1771-81, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471359

ABSTRACT

The cardiac L-type calcium current (I(Ca,L)) is an important regulator of myocardial contractility. It is activated by sympathetic stimulation and inhibited by parasympathetic activity via muscarinic acetylcholine receptors. Muscarinic inhibition of I(Ca,L) occurs via activation of pertussis toxin (PTX)-sensitive heterotrimeric G-proteins. Although recent studies have shown that expression of G(oalpha) is important for this effect in adult mouse ventricular cells, two other PTX-sensitive G-proteins (G(i2) and G(i3)) are also expressed in cardiocytes and are activated. Their role in the regulation of I(Ca,L) has not been examined. In addition, it is not known whether nodal/atrial cardiac cells use the same G-proteins. We show that gene inactivation of each of the three PTX-sensitive Galpha-proteins (alpha(i2), alpha(i3), and alpha(o)) affects muscarinic inhibition of cardiac I(Ca,L) in embryonic stem (ES) cell-derived cardiocytes. Inactivation of either alpha(i2) or alpha(i3) markedly slows the time course of muscarinic inhibition of I(Ca,L), and in cells where both alpha(i2) and alpha(i3) are inactivated the effects are not additive. We also establish an essential role for alpha(o)in this atrial/nodal-like cardiocyte system and show that alpha(o)acts proximal to NO generation. NO generation plays a critical role in I(Ca,L) regulation since the nitric oxide synthase (NOS) antagonist, l -NMMA, blocked the inhibition of I(Ca,L) in WT and in alpha(i2)/alpha(i3)-null cells. In WT cells, the NO generating agent SIN-1 inhibited I(Ca,L) and the addition of carbachol resulted in faster inhibition, suggesting that pathways in addition to NO are also activated. This study shows that alpha(i2) and alpha(i3) play a critical role in the normal inhibition of cardiocyte I(Ca,L). Thus, all muscarinic receptor activated G-proteins (G(i2), G(i3) and G(o)) are necessary for normal inhibition and act through both NO and non-NO signaling pathways.


Subject(s)
Calcium Channels/physiology , GTP-Binding Proteins/metabolism , Heart/drug effects , Isoproterenol/pharmacology , Myocardium/cytology , Pertussis Toxin , Receptors, Muscarinic/physiology , Virulence Factors, Bordetella/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium Channels/drug effects , Calcium Channels, L-Type , Carbachol/pharmacology , Cell Line , GTP-Binding Proteins/deficiency , Heart Ventricles , Mice , Models, Cardiovascular , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Muscarinic Antagonists/pharmacology , Nifedipine/pharmacology , Nitric Oxide Donors/pharmacology , Stem Cells/cytology , omega-N-Methylarginine/pharmacology
14.
J Biol Chem ; 274(32): 22739-46, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10428857

ABSTRACT

Calcium is a major component of the mineral phase of bone and serves as a key intracellular second messenger. Postnatally, all bodily calcium must be absorbed from the diet through the intestine. Here we report the properties of a calcium transport protein (CaT1) cloned from rat duodenum using an expression cloning strategy in Xenopus laevis oocytes, which likely plays a key role in the intestinal uptake of calcium. CaT1 shows homology (75% amino acid sequence identity) to the apical calcium channel ECaC recently cloned from vitamin D-responsive cells of rabbit kidney and is structurally related to the capsaicin receptor and the TRP family of ion channels. Based on Northern analysis of rat tissues, a 3-kilobase CaT1 transcript is present in rat duodenum, proximal jejunum, cecum, and colon, and a 6.5-kilobase transcript is present in brain, thymus, and adrenal gland. In situ hybridization revealed strong CaT1 mRNA expression in enterocytes of duodenum, proximal jejunum, and cecum. No signals were detected in kidney, heart, liver, lung, spleen, and skeletal muscle. When expressed in Xenopus oocytes, CaT1 mediates saturable Ca(2+) uptake with a Michaelis constant of 0.44 mM. Transport of Ca(2+) by CaT1 is electrogenic, voltage-dependent, and exhibits a charge/Ca(2+) uptake ratio close to 2:1, indicating that CaT1-mediated Ca(2+) influx is not coupled to other ions. CaT1 activity is pH-sensitive, exhibiting significant inhibition by low pH. CaT1 is also permeant to Sr(2+) and Ba(2+) (but not Mg(2+)), although the currents evoked by Sr(2+) and Ba(2+) are much smaller than those evoked by Ca(2+). The trivalent cations Gd(3+) and La(3+) and the divalent cations Cu(2+), Pb(2+), Cd(2+), Co(2+), and Ni(2+) (each at 100 microM) do not evoke currents themselves, but inhibit CaT1-mediated Ca(2+) transport. Fe(3+), Fe(2+), Mn(2+), and Zn(2+) have no significant effects at 100 microM on CaT1-mediated Ca(2+) transport. CaT1 mRNA levels are not responsive to 1,25-dihydroxyvitamin D(3) administration or to calcium deficiency. Our studies strongly suggest that CaT1 provides the principal mechanism for Ca(2+) entry into enterocytes as part of the transcellular pathway of calcium absorption in the intestine.


Subject(s)
Calcium Channels/genetics , Calcium, Dietary/metabolism , Calcium/metabolism , Intestinal Absorption/genetics , Amino Acid Sequence , Animals , Calcitriol/pharmacology , Calcium/deficiency , Calcium Channels/metabolism , Cloning, Molecular/methods , DNA, Complementary/genetics , Electric Conductivity , Electrophysiology , Gene Expression , Gene Library , Models, Molecular , Molecular Sequence Data , Patch-Clamp Techniques , RNA, Messenger/isolation & purification , Rats , TRPV Cation Channels , Xenopus laevis
15.
J Neurochem ; 72(5): 1915-22, 1999 May.
Article in English | MEDLINE | ID: mdl-10217268

ABSTRACT

The calcium-sensing receptor (CaR) is a G protein-coupled receptor that "senses" extracellular calcium ions (Ca2+o) as an extracellular first messenger. In this report, we have shown that the CaR is expressed in primary cultures of microglial cells derived from rat brain as assessed by RT-PCR using four CaR-specific primer pairs followed by sequencing of the amplified products, by northern blot analysis using a CaR-specific probe, as well as by immunocytochemistry and western analysis utilizing a specific polyclonal anti-CaR antiserum. In addition, raising Ca2+o from 0.75 to 3.0 mM or addition of the polycationic CaR agonist neomycin or a "calcimimetic" CaR activator (R-467; NPS Pharmaceuticals) increased the open state probability (Po) of a Ca(+)-activated K+ channel having a unitary conductance of 84+/-4 pS, indicating that the channel is modulated by the CaR. Therefore, our data strongly suggest that a functional CaR is expressed in cultured rat microglia, similar to that in parathyroid gland and kidney, which could potentially play an important role(s) in regulating microglial function.


Subject(s)
Extracellular Space/metabolism , Microglia/metabolism , Potassium Channels/metabolism , Receptors, Cell Surface/physiology , Animals , Biomarkers , Blotting, Western , Calcium/physiology , Gene Expression/physiology , Microglia/physiology , Rats , Receptors, Calcium-Sensing , Receptors, Cell Surface/agonists , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Glia ; 26(1): 64-72, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10088673

ABSTRACT

An extracellular calcium (Ca2+o)-sensing receptor (CaR) plays crucial roles in maintaining systemic calcium homeostasis. The CaR is also expressed in other cells uninvolved in systemic mineral ion homeostasis, including keratinocytes, fibroblasts, and neurons. In brain the CaR is widely distributed, being particularly abundant in neurons in subfornical organ, cingulate cortex, hippocampus, and cerebellum. It is also present in fiber tracts in rat brain, presumably in oligodendroglia and in cultured rat oligodendrocytes, suggesting that the CaR modulates the function of nonneuronal cells within brain. In this report, we show functional CaR expression in a human astrocytoma cell line (U87). Reverse transcription-polymerase chain reaction (RT-PCR) amplified a product from U87 cell RNA exhibiting >98% homology with the human CaR. Northern blot revealed a 5.5 kb transcript, similar to the principal transcript in human parathyroid, and a smaller 2.4 kb transcript. U87 cells expressed CaR protein as assessed by immunocytochemistry and Western blot using an affinity-purified, anti-CaR antiserum. Patch clamp analysis in the cell-attached mode revealed that raising Ca2+o from 0.75 to 1.75 or 2.75 mM produced approximately threefold increases in the open state probability (Po) of an outward K+ channel with a conductance of approximately 88 pS. A specific "calcimimetic" CaR activator, R-467 (0.5 microM), activated this K+ channel similarly, while its less active stereoisomer, S-467, did not. Thus U87 astrocytoma cells express both CaR mRNA and protein, and the receptor activates an outward K+ channel previously suggested to be involved in membrane polarization and cellular excitability.


Subject(s)
Astrocytoma/physiopathology , Calcium Channels/physiology , Potassium Channels/physiology , Transcription, Genetic , Animals , Blotting, Northern , Brain Neoplasms/physiopathology , Calcium/pharmacology , Calcium/physiology , Calcium Channels/genetics , Humans , Ion Channel Gating , Membrane Potentials/drug effects , Membrane Potentials/physiology , Oligodendroglia/physiology , Patch-Clamp Techniques , Potassium Channels/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stereoisomerism , Tumor Cells, Cultured
18.
Brain Res ; 851(1-2): 116-24, 1999 Dec 18.
Article in English | MEDLINE | ID: mdl-10642834

ABSTRACT

A receptor for extracellular calcium ions (Ca2+o), cloned from parathyroid gland, serves a critical function in Ca2+o homeostasis by regulating PTH release via "sensing" of its physiological agonist, Ca2+o. Its cloning from rat striatum revealed that the extracellular calcium-sensing receptor (CaR) could be involved in sensing ambient Ca2+o within the brain, where Ca2+ plays key roles in virtually all aspects of central nervous system (CNS) function. The CaR is expressed in neurons, oligodendrocytes, microglia and the human astrocytoma cell line, U87 where its functions include control of cellular proliferation and modulation of ion channels, such as outward K+ channels and nonselective cation channels (NCC). In this report, we have shown that the CaR is expressed in U373 cells as assessed by RT-PCR using CaR-specific primers followed by sequencing of the amplified products, by Northern blot analysis using a CaR-specific probe as well as by Western analysis utilizing a specific polyclonal anti-CaR antiserum. Furthermore, agents known to activate the cloned CaR induce increases in cellular proliferation and the open probability of an NCC. Thus our study strongly suggests that elevated levels of Ca2+o, acting via the CaR, activate an NCC that could contribute to the associated CaR-induced stimulation of proliferation.


Subject(s)
Astrocytoma/metabolism , Calcium/metabolism , Glioblastoma/metabolism , Ion Pumps/metabolism , Receptors, Cell Surface/physiology , Animals , Calcium/pharmacology , Cell Division/physiology , Humans , Ion Pumps/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Middle Aged , Rats , Receptors, Calcium-Sensing , Receptors, Cell Surface/agonists , Tumor Cells, Cultured
19.
Glia ; 24(4): 449-58, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9814825

ABSTRACT

A G protein-coupled, extracellular calcium (Ca(0)2+)-sensing receptor (CaR) cloned from parathyroid, kidney, and brain plays crucial roles in systemic calcium metabolism. In brain, the CaR is located in nerve terminals as well as in fiber tracts, where it may be expressed in glia. Moreover, there is Ca2+- and K+-dependent communication between axons and oligodendroglia. To investigate further the potential role of the CaR in oligodendroglia, we studied expression of CaR mRNA and protein as well as the effects of CaR agonists on cellular proliferation and Ca2+-activated K+ channel activity in immature rat oligodendrocytes in primary culture. Reverse transcriptase polymerase chain reaction and sequencing of CaR transcripts from oligodendrocytes revealed >99% sequence identity with the rat kidney CaR. Northern analysis demonstrated 7.5 and 4.1 kb transcripts in oligodendrocytes, similar to those in rat parathyroid and kidney, while Western analysis and immunocytochemistry with CaR-specific antisera showed the presence of CaR protein. Immunocytochemically, the CaR was colocalized with galactocerebroside in the cultured oligodendrocytes. Raising Ca(0)2+ from 1.8 to 4.8 mM or addition of the polycationic CaR agonist neomycin (300 microM) modestly but significantly increased [3H]-thymidine incorporation into oligodendrocytes. Elevating Ca(0)2+ from 0.75 to 3.0 mM or addition of 100 microM neomycin also produced 2-2.5-fold increases in the open state probability (Po) of an outward K+ channel with a unitary conductance of 88+/-5 pS. Taken together, our data show that the CaR is expressed in immature oligodendrocytes and may be functionally linked to cellular proliferation and an outward K+ channel potentially contributing to local ionic homeostasis in the vicinity of oligodendroglia.


Subject(s)
Calcium/metabolism , Oligodendroglia/metabolism , Potassium Channels/metabolism , Receptors, Cell Surface/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Division , Extracellular Space/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression , Neomycin/pharmacology , Polymerase Chain Reaction/methods , Rats , Rats, Wistar , Receptors, Calcium-Sensing , Receptors, Cell Surface/genetics
20.
Recent Prog Horm Res ; 53: 257-80; discussion 280-1, 1998.
Article in English | MEDLINE | ID: mdl-9769711

ABSTRACT

The ability of parathyroid cells to recognize and respond to (i.e., "sense") small changes in the extracellular Ca2+ concentration (Ca2+o) plays a crucial role in mineral ion homeostasis. Expression cloning in Xenopus laevis oocytes enabled isolation of a cDNA coding for the bovine parathyroid CaR. CaRs were later isolated from human parathyroid and kidney, rat kidney, brain and C-cell, rabbit kidney, and chicken parathyroid. All are tissue and species homologs of the same ancestral gene. The predicted CaR protein has a large extracellular amino-terminus, which binds polycationic CaR agonists; a central core with seven membrane-spanning helices, documenting that it is a G protein-coupled receptor; and an approximately 200 amino acid carboxyl-terminal tail. The CaR is highly expressed in parathyroid and C-cells, along almost the entire nephron and gastrointestinal (GI) tract and within numerous regions of the brain, particularly hippocampus, cerebellum, and hypothalamus. The CaR's physiological importance has been documented by the identification of hyper- and hypocalcemic syndromes due to inactivating or activating CaR mutations, respectively. Familial hypocalciuric hypercalcemia (FHH) and neonatal severe hyperparathyroidism (NSHPT) are caused by loss-of-function CaR mutations producing Ca2+o "resistance," while autosomal dominant hypocalcemia is the result of activating mutations rendering CaRs overly sensitive to Ca2+o. In addition to showing altered parathyroid responsiveness to Ca2+o, patients with FHH reabsorb too much urinary Ca2+ and Mg2+ at a given Ca2+o, while those with autosomal dominant hypocalcemia excrete too much, illustrating the CaR's key role in renal handling of divalent cations. Recent in vitro data suggest that the CaR directly regulates renal water handling in the collecting duct. Indeed, patients with FHH concentrate their urine normally, despite their hypercalcemia, while those with autosomal dominant hypocalcemia can exhibit impaired urinary concentration at normal or even low Ca2+o, suggesting that the CaR enables coordination of renal calcium and water handling. In addition to serving these "homeostatic" roles, the CaR likely also enables Ca2+o to serve additional roles as an extracellular messenger. The receptor regulates key Ca2+ and K(+)-permeable ion channels in hippocampal and other brain cells and likely senses local changes in Ca2+o within the brain microenvironment accompanying neuronal activation. It is also present in and regulates ion channels in lens epithelial cells, potentially playing some role in cataract development in hypoparathyroid patients. In keratinocytes and epithelial cells of the gastrointestinal tract, in contrast, the CaR may regulate cellular proliferation and differentiation, processes known to be modulated by Ca2+o in these cell types. Thus, in addition to sensing and regulating systemic Ca2+o, the CaR likely enables Ca2+o to act as a local signal for cells within specific microenvironments, such as the brain or eye.


Subject(s)
Calcium Signaling/physiology , Receptors, Cell Surface/physiology , Amino Acid Sequence , Animals , Molecular Sequence Data , Receptors, Calcium-Sensing
SELECTION OF CITATIONS
SEARCH DETAIL
...