Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
Add more filters










Publication year range
1.
Oncogene ; 33(18): 2307-16, 2014 May 01.
Article in English | MEDLINE | ID: mdl-23686305

ABSTRACT

Signals from the tumor microenvironment trigger cancer cells to adopt an invasive phenotype through epithelial-mesenchymal transition (EMT). Relatively little is known regarding key signal transduction pathways that serve as cytosolic bridges between cell surface receptors and nuclear transcription factors to induce EMT. A better understanding of these early EMT events may identify potential targets for the control of metastasis. One rapid intracellular signaling pathway that has not yet been explored during EMT induction is calcium. Here we show that stimuli used to induce EMT produce a transient increase in cytosolic calcium levels in human breast cancer cells. Attenuation of the calcium signal by intracellular calcium chelation significantly reduced epidermal growth factor (EGF)- and hypoxia-induced EMT. Intracellular calcium chelation also inhibited EGF-induced activation of signal transducer and activator of transcription 3 (STAT3), while preserving other signal transduction pathways such as Akt and extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation. To identify calcium-permeable channels that may regulate EMT induction in breast cancer cells, we performed a targeted siRNA-based screen. We found that transient receptor potential-melastatin-like 7 (TRPM7) channel expression regulated EGF-induced STAT3 phosphorylation and expression of the EMT marker vimentin. Although intracellular calcium chelation almost completely blocked the induction of many EMT markers, including vimentin, Twist and N-cadherin, the effect of TRPM7 silencing was specific for vimentin protein expression and STAT3 phosphorylation. These results indicate that TRPM7 is a partial regulator of EMT in breast cancer cells, and that other calcium-permeable ion channels are also involved in calcium-dependent EMT induction. In summary, this work establishes an important role for the intracellular calcium signal in the induction of EMT in human breast cancer cells. Manipulation of calcium-signaling pathways controlling EMT induction in cancer cells may therefore be an important therapeutic strategy for preventing metastases.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium Signaling , Calcium/metabolism , Epithelial-Mesenchymal Transition/physiology , Cell Hypoxia , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Serine-Threonine Kinases , RNA, Small Interfering/genetics , STAT3 Transcription Factor/metabolism , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Vimentin/biosynthesis
2.
Handb Exp Pharmacol ; (179): 593-614, 2007.
Article in English | MEDLINE | ID: mdl-17217081

ABSTRACT

The canonical transient receptor potential (TRPC) cation channels are mammalian homologs of the photoreceptor channel TRP in Drosophila melanogaster. All seven TRPCs (TRPC1 through TRPC7) can be activated through Gq/11 receptors or receptor tyrosine kinase (RTK) by mechanisms downstream of phospholipase C. The last decade saw a rapidly growing interest in understanding the role of TRPC channels in calcium entry pathways as well as in understanding the signal(s) responsible for TRPC activation. TRPC channels have been proposed to be activated by a variety of signals including store depletion, membrane lipids, and vesicular insertion into the plasma membrane. Here we discuss recent developments in the mode of activation as well as the pharmacological and electrophysiological properties of this important and ubiquitous family of cation channels.


Subject(s)
TRPC Cation Channels/physiology , Type C Phospholipases/physiology , Animals , Biotransformation/physiology , Calcium/metabolism , Humans
3.
Proc Natl Acad Sci U S A ; 98(20): 11777-82, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11553786

ABSTRACT

Mammalian Trp proteins are candidates for plasma membrane calcium channels regulated by receptor activation or by intracellular calcium store depletion [capacitative calcium entry (CCE)]. One extensively investigated member of the Trp family, the human Trp3 (hTrp3), behaves as a receptor-activated, calcium-permeable, nonselective cation channel when expressed in cell lines and does not appear to be activated by store depletion. Nonetheless, there is good evidence that Trp3 can be regulated by interacting with inositol trisphosphate receptors (IP(3)Rs), reminiscent of the conformational coupling mode of CCE. To investigate the role of Trp3 in CCE, and its regulation by IP(3)R, we transiently expressed hTrp3 in the wild-type DT40 chicken B lymphocyte cell line and its variant lacking IP(3)R. Expression of hTrp3 in either wild-type or IP(3)R-knockout cells did not increase basal membrane permeability, but resulted in a substantially greater divalent cation entry after thapsigargin-induced store depletion. This hTrp3-dependent divalent cation entry was significantly greater in the wild type than in IP(3)R-knockout cells. Thus, it appears that in this cell line, hTrp3 forms channels that are store-operated by both IP(3)R-dependent and IP(3)R-independent mechanisms. Trp3, or one of its structural relatives, is a candidate for the store-operated, nonselective cation channels observed in smooth muscle cells and other cell types.


Subject(s)
B-Lymphocytes/physiology , Calcium Channels/physiology , Ion Channels/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , B-Lymphocytes/drug effects , Barium/metabolism , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Chickens , Cloning, Molecular , DNA Primers , Fura-2 , Genetic Vectors , Humans , Inositol 1,4,5-Trisphosphate Receptors , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels , Thapsigargin/pharmacology , Transcription, Genetic , Transfection
4.
J Cell Sci ; 114(Pt 12): 2223-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11493662

ABSTRACT

Capacitative Ca(2+) entry involves the regulation of plasma membrane Ca(2+) channels by the filling state of intracellular Ca(2+) stores in the endoplasmic reticulum (ER). Several theories have been advanced regarding the mechanism by which the stores communicate with the plasma membrane. One such mechanism, supported by recent findings, is conformational coupling: inositol 1,4,5-trisphosphate (Ins(1,4,5)P(3)) receptors in the ER may sense the fall in Ca(2+) levels through Ca(2+)-binding sites on their lumenal domains, and convey this conformational information directly by physically interacting with Ca(2+) channels in the plasma membrane. In support of this idea, in some cell types, store-operated channels in excised membrane patches appear to depend on the presence of both Ins(1,4,5)P(3) and Ins(1,4,5)P(3) receptors for activity; in addition, inhibitors of Ins(1,4,5)P(3) production that either block phospholipase C or inhibit phosphatidylinositol 4-kinase can block capacitative Ca(2+) entry. However, the electrophysiological current underlying capacitative Ca(2+) entry is not blocked by an Ins(1,4,5)P(3) receptor antagonist, and the blocking effects of a phospholipase C inhibitor are not reversed by the intracellular application of Ins(1,4,5)P(3). Furthermore, cells whose Ins(1,4,5)P(3) receptor genes have been disrupted can nevertheless maintain their capability to activate capacitative Ca(2+) entry channels in response to store depletion. A tentative conclusion is that multiple mechanisms for signaling capacitative Ca(2+) entry may exist, and involve conformational coupling in some cell types and perhaps a diffusible signal in others.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Ion Channel Gating , Animals , Electric Conductivity , Endoplasmic Reticulum/metabolism , Exocytosis , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Models, Biological , Receptors, Cytoplasmic and Nuclear/metabolism
6.
J Biol Chem ; 276(23): 20186-9, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11274150

ABSTRACT

In nonexcitable cells, the predominant mechanism for regulated entry of Ca(2+) is capacitative calcium entry, whereby depletion of intracellular Ca(2+) stores signals the activation of plasma membrane calcium channels. A number of other regulated Ca(2+) entry pathways occur in specific cell types, however, and it is not know to what degree the different pathways interact when present in the same cell. In this study, we have examined the interaction between capacitative calcium entry and arachidonic acid-activated calcium entry, which co-exist in HEK293 cells. These two pathways exhibit mutual antagonism. That is, capacitative calcium entry is potently inhibited by arachidonic acid, and arachidonic acid-activated entry is inhibited by the pre-activation of capacitative calcium entry with thapsigargin. In the latter case, the inhibition does not seem to result from a direct action of thapsigargin, inhibition of endoplasmic reticulum Ca(2+) pumps, depletion of Ca(2+) stores, or entry of Ca(2+) through capacitative calcium entry channels. Rather, it seems that a discrete step in the pathway signaling capacitative calcium entry interacts with and inhibits the arachidonic acid pathway. The findings reveal a novel process of mutual antagonism between two distinct calcium entry pathways. This mutual antagonism may provide an important protective mechanism for the cell, guarding against toxic Ca(2+) overload.


Subject(s)
Arachidonic Acid/metabolism , Calcium/antagonists & inhibitors , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , Humans , Thapsigargin/pharmacology
7.
J Biol Chem ; 276(19): 15945-52, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278938

ABSTRACT

We investigated the putative roles of phospholipase C, polyphosphoinositides, and inositol 1,4,5-trisphosphate (IP(3)) in capacitative calcium entry and calcium release-activated calcium current (I(crac)) in lacrimal acinar cells, rat basophilic leukemia cells, and DT40 B-lymphocytes. Inhibition of phospholipase C with blocked calcium entry and I(crac) activation whether in response to a phospholipase C-coupled agonist or to calcium store depletion with thapsigargin. Run-down of cellular polyphosphoinositides by concentrations of wortmannin that block phosphatidylinositol 4-kinase completely blocked calcium entry and I(crac). The membrane-permeant IP(3) receptor inhibitor, 2-aminoethoxydiphenyl borane, blocked both capacitative calcium entry and I(crac). However, it is likely that 2-aminoethoxydiphenyl borane does not inhibit through an action on the IP(3) receptor because the drug was equally effective in wild-type DT40 B-cells and in DT40 B-cells whose genes for all three IP(3) receptors had been disrupted. Intracellular application of another potent IP(3) receptor antagonist, heparin, failed to inhibit activation of I(crac). Finally, the inhibition of I(crac) activation by or wortmannin was not reversed or prevented by direct intracellular application of IP(3). These findings indicate a requirement for phospholipase C and for polyphosphoinositides for activation of capacitative calcium entry. However, the results call into question the previously suggested roles of IP(3) and IP(3) receptor in this mechanism, at least in these particular cell types.


Subject(s)
Calcium/physiology , Inositol 1,4,5-Trisphosphate/metabolism , Lacrimal Apparatus/metabolism , Phosphatidylinositol Phosphates/metabolism , Type C Phospholipases/metabolism , Androstadienes/pharmacology , Animals , B-Lymphocytes , Boron Compounds/pharmacology , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Channels/physiology , Cell Line, Transformed , Cells, Cultured , Chickens , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Heparin/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Kinetics , Lacrimal Apparatus/cytology , Lacrimal Apparatus/drug effects , Leukemia, Basophilic, Acute , Mice , Pyrrolidinones/pharmacology , Rats , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Thapsigargin/pharmacology , Tumor Cells, Cultured , Wortmannin
8.
J Biol Chem ; 276(2): 1063-70, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11042187

ABSTRACT

The regulation of store-operated, calcium-selective channels in the plasma membrane of rat basophilic leukemia cells (RBL-2H3 m1), an immortalized mucosal mast cell line, was studied at the single-channel level with the patch clamp technique by removing divalent cations from both sides of the membrane. The activity of the single channels in excised patches could be modulated by Ca(2+), Mg(2+), and pH. The maximal activation of these channels by divalent cation-free conditions occurred independently of depletion of intracellular Ca(2+) stores, whether in excised patches or in whole cell mode. Yet, a number of points of evidence establish these single-channel openings as amplified store-operated channel events. Specifically, (i) the single channels are exquisitely sensitive to inhibition by intracellular Ca(2+), and (ii) both the store-operated current and the single-channel openings are completely blocked by the capacitative calcium entry blocker, 2-aminoethoxydiphenyl borane. In addition, in Jurkat T cells single-channel openings with lower open probability have been observed in the whole cell mode with intracellular Mg(2+) present (Kerschbaum, H. H., and Cahalan, M. D. (1999) Science 283, 836-839), and in RBL-2H3 m1 cells a current with similar properties is activated by store depletion.


Subject(s)
Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Calcium/pharmacology , Cell Membrane/physiology , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Hydrogen-Ion Concentration , Leukemia, Basophilic, Acute , Magnesium/pharmacology , Patch-Clamp Techniques , Rats , Ryanodine Receptor Calcium Release Channel/drug effects , Solutions , Tumor Cells, Cultured
9.
J Biol Chem ; 276(8): 5613-21, 2001 Feb 23.
Article in English | MEDLINE | ID: mdl-11096083

ABSTRACT

We have investigated the signaling pathways underlying muscarinic receptor-induced calcium oscillations in human embryonic kidney (HEK293) cells. Activation of muscarinic receptors with a maximal concentration of carbachol (100 microm) induced a biphasic rise in cytoplasmic calcium ([Ca2+]i) comprised of release of Ca2+ from intracellular stores and influx of Ca2+ from the extracellular space. A lower concentration of carbachol (5 microm) induced repetitive [Ca2+]i spikes or oscillations, the continuation of which was dependent on extracellular Ca2+. The entry of Ca2+ with 100 microm carbachol and with the sarcoplasmic-endoplasmic reticulum calcium ATPase inhibitor, thapsigargin, was completely blocked by 1 microm Gd3+, as well as 30-100 microm concentrations of the membrane-permeant inositol 1,4,5-trisphosphate receptor inhibitor, 2-aminoethyoxydiphenyl borane (2-APB). Sensitivity to these inhibitors is indicative of capacitative calcium entry. Arachidonic acid, a candidate signal for Ca2+ entry associated with [Ca2+]i oscillations in HEK293 cells, induced entry that was inhibited only by much higher concentrations of Gd3+ and was unaffected by 100 microm 2-APB. Like arachidonic acid-induced entry, the entry associated with [Ca2)]i oscillations was insensitive to inhibition by Gd3+ but was completely blocked by 100 microm 2-APB. These findings indicate that the signaling pathway responsible for the Ca2+) entry driving [Ca2+]i oscillations in HEK293 cells is more complex than originally thought, and may involve neither capacitative calcium entry nor a role for PLA2 and arachidonic acid.


Subject(s)
Calcium Signaling , Receptors, Muscarinic/metabolism , Arachidonic Acid/pharmacology , Biological Transport , Boron Compounds/pharmacology , Carbachol/pharmacology , Drug Interactions , Gadolinium/pharmacology , Humans , Thapsigargin/pharmacology
10.
Mol Interv ; 1(2): 84-94, 2001 Jun.
Article in English | MEDLINE | ID: mdl-14993328

ABSTRACT

The versatility of Ca(2+) as a messenger in multiple signaling events requires that the concentration of calcium ions within the cytoplasm be highly regulated. In particular, the release of calcium from intracellular stores must often be linked to calcium influx across the cell membrane. Capacitative calcium entry, whereby the depletion of intracellular Ca(2+) stores induces the influx of extracellular calcium, is a crucial element of concerted calcium signaling. Investigations into the phenomenon are contributing to a new appreciation for the organized cytoplasmic framework that supports calcium signaling.


Subject(s)
Calcium Signaling , Calcium/antagonists & inhibitors , Cell Membrane/metabolism , Ion Transport/physiology , Pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Humans , Models, Biological
11.
Biochem J ; 351 Pt 3: 735-46, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11042129

ABSTRACT

Mammalian homologues of the Drosophila transient receptor potential (TRP) protein have been proposed to function as ion channels, and in some cases as store-operated or capacitative calcium entry channels. However, for each of the mammalian TRP proteins, different laboratories have reported distinct modes of cellular regulation. In the present study we describe the cloning and functional expression of the human form of TRP4 (hTRP4), and compare its activity with another well studied protein, hTRP3. When hTRP4 was transiently expressed in human embryonic kidney (HEK)-293 cells, basal bivalent cation permeability (barium) was increased. Whole-cell patch-clamp studies of hTRP4 expressed in Chinese hamster ovary cells revealed a constitutively active non-selective cation current which probably underlies the increased bivalent cation entry. Barium entry into hTRP4-transfected HEK-293 cells was not further increased by phospholipase C (PLC)-linked receptor activation, by intracellular calcium store depletion with thapsigargin, or by a synthetic diacylglycerol, 1-oleoyl-2-acetyl-sn-glycerol (OAG). In contrast, transient expression of hTRP3 resulted in a bivalent cation influx that was markedly increased by PLC-linked receptor activation and by OAG, but not by thapsigargin. Despite the apparent differences in regulation of these two putative channel proteins, green fluorescent protein fusions of both molecules localized similarly to the plasma-membrane, notably in discrete punctate regions suggestive of specialized signalling complexes. Our findings indicate that while both hTRP4 and hTRP3 can apparently function as cation channels, their putative roles as components of capacitative calcium entry channels are not readily demonstrable by examining their behaviour when exogenously expressed in cells.


Subject(s)
Calcium Channels/genetics , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium Channels/physiology , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 13 , Cloning, Molecular , Cricetinae , DNA Primers , DNA, Complementary , Humans , Molecular Sequence Data , Patch-Clamp Techniques , Protein Conformation , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels
12.
Cell Mol Life Sci ; 57(8-9): 1272-86, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11028918

ABSTRACT

This review discusses multiple ways in which the endoplasmic reticulum participates in and is influenced by signal transduction pathways. The endoplasmic reticulum provides a Ca2+ store that can be mobilized either by calcium-induced calcium release or by the diffusible messenger inositol 1,4,5-trisphosphate. Depletion of endoplasmic reticulum Ca2+ stores provides a signal that activates surface membrane Ca2+ channels, a process known as capacitative calcium entry. Depletion of endoplasmic reticulum stores can also signal long-term cellular responses such as gene expression and programmed cell death or apoptosis. In addition to serving as a source of cellular signals, the endoplasmic reticulum is also functionally and structurally modified by the Ca2+ and protein kinase C pathways. Elevated cytoplasmic Ca2+ causes a rearrangement and fragmentation of endoplasmic reticulum membranes. Protein kinase C activation reduces the storage capacity of the endoplasmic reticulum Ca2+ pool. In some cell types, protein kinase C inhibits capacitative calcium entry. Protein kinase C activation also protects the endoplasmic reticulum from the structural effects of high cytoplasmic Ca2+. The emerging view is one of a complex network of pathways through which the endoplasmic reticulum and the Ca2+ and protein kinase C signaling pathways interact at various levels regulating cellular structure and function.


Subject(s)
Calcium Signaling/physiology , Cell Membrane/physiology , Endoplasmic Reticulum/physiology , Animals , Calcium/metabolism , Humans
14.
Cell Calcium ; 27(3): 175-85, 2000 Mar.
Article in English | MEDLINE | ID: mdl-11007130

ABSTRACT

In human embryonic kidney (HEK) cells stably transfected with green fluorescent protein targeted to the endoplasmic reticulum (ER), elevation of intracellular Ca2+ ([Ca2+]i) altered ER morphology, making it appear punctate. Electron microscopy revealed that these punctate structures represented circular and branched rearrangements of the endoplasmic reticulum, but did not involve obvious swelling or pathological fragmentation. Activation of protein kinase C with phorbol 12-myristate 13-acetate (PMA), prevented the effects of ionomycin on ER structure without affecting the elevation of [Ca2+]i. These results suggest that protein kinase C activation alters cytoplasmic or ER components underlying the effects of high [Ca2+]i on ER structure. Treatment of HEK cells with PMA also reduced the size of the thapsigargin-sensitive Ca2+ pool and inhibited Ca2+ entry in response to thapsigargin. Thus, protein kinase C activation has multiple actions on the calcium storage and signalling function of the endoplasmic reticulum in HEK cells: (1) reduced intracellular Ca2+ storage capacity, (2) inhibition of capacitative Ca2+ entry, and (3) protection of the endoplasmic reticulum against the effects of high [Ca2+]i.


Subject(s)
Calcium Signaling , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Protein Kinase C/metabolism , Cell Line , Cytophotometry , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Fura-2/metabolism , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Kidney , Methacholine Chloride/pharmacology , Microscopy, Confocal , Microscopy, Electron , Muscarinic Agonists/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Transfection
15.
J Biol Chem ; 275(39): 30586-96, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-10859318

ABSTRACT

Jurkat cells undergo apoptosis in response to anti-Fas antibody through a caspase-dependent death cascade in which calcium signaling has been implicated. We have now evaluated the role of calcium during this death cascade at the single cell level in real time utilizing flow cytometric analysis and confocal microscopy. Fluo-3 and propidium iodide were employed to evaluate calcium fluxes and to discriminate between viable and non-viable cells, respectively. Anti-Fas treatment of Jurkat cells resulted in a sustained increase in intracellular calcium commencing between 1 and 2 h after treatment and persisting until subsequent loss of cell membrane integrity. The significance of this rise in calcium was evaluated by buffering intracellular calcium with BAPTA and/or removing calcium from the extracellular medium and monitoring the effects of these manipulations on calcium signaling and components of the apoptotic process. Complete inhibition of the anti-Fas induced rise in intracellular calcium required both chelation of [Ca(2+)](i) and removal of extracellular calcium. Interestingly, this condition did not abrogate several events in Fas-induced apoptosis including cell shrinkage, mitochondrial depolarization, annexin binding, caspase activation, and nuclear poly(A)DP-ribose polymerase cleavage. Furthermore, calcium-free conditions in the absence of anti-Fas antibody weakly induced these apoptotic components. In marked contrast, calcium depletion did not induce DNA degradation in control cells, and inhibited apoptotic DNA degradation in response to anti-Fas. These data support the concept that the rise in intracellular calcium is not a necessary component for the early signal transduction pathways in anti-Fas-induced apoptosis in Jurkat cells, but rather is necessary for the final degradation of chromatin via nuclease activation.


Subject(s)
Apoptosis , Calcium/metabolism , DNA, Neoplasm/metabolism , fas Receptor/metabolism , Biological Transport , Caspase 3 , Caspases/metabolism , Cell Nucleus/ultrastructure , Cell Size , Chelating Agents/pharmacology , Chromatin/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Flow Cytometry , Humans , Jurkat Cells , Membrane Lipids/analysis , Membrane Potentials/drug effects , Microscopy, Confocal , Mitochondria/physiology , Phosphatidylserines/pharmacology , Signal Transduction
17.
J Biol Chem ; 274(46): 32881-8, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10551852

ABSTRACT

We examined the activation and regulation of calcium release-activated calcium current (I(crac)) in RBL-1 cells in response to various Ca(2+) store-depleting agents. With [Ca(2+)](i) strongly buffered to 100 nM, I(crac) was activated by ionomycin, thapsigargin, inositol 1,4,5-trisphosphate (IP(3)), and two metabolically stable IP(3) receptor agonists, adenophostin A and L-alpha-glycerophospho-D-myoinositol-4,5-bisphosphate (GPIP(2)). With minimal [Ca(2+)](i) buffering, with [Ca(2+)](i) free to fluctuate I(crac) was activated by ionomycin, thapsigargin, and by the potent IP(3) receptor agonist, adenophostin A, but not by GPIP(2) or IP(3) itself. Likewise, when [Ca(2+)](i) was strongly buffered to 500 nM, ionomycin, thapsigargin, and adenophostin A did and GPIP(2) and IP(3) did not activate detectable I(crac). However, with minimal [Ca(2+)](i) buffering, or with [Ca(2+)](i) buffered to 500 nM, GPIP(2) was able to fully activate detectable I(crac) if uptake of Ca(2+) intracellular stores was first inhibited. Our findings suggest that when IP(3) activates the IP(3) receptor, the resulting influx of Ca(2+) quickly inactivates the receptor, and Ca(2+) is re-accumulated at sites that regulate I(crac). Adenophostin A, by virtue of its high receptor affinity, is resistant to this inactivation. Comparison of thapsigargin-releasable Ca(2+) pools following activation by different IP(3) receptor agonists indicates that the critical regulatory pool of Ca(2+) may be very small in comparison to the total IP(3)-sensitive component of the endoplasmic reticulum. These findings reveal new and important roles for IP(3) receptors located on discrete IP(3)-sensitive Ca(2+) pools in calcium feedback regulation of I(crac) and capacitative calcium entry.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Feedback , Fluorescence , Fura-2/chemistry , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Inositol Phosphates/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Patch-Clamp Techniques , Rats , Thapsigargin/pharmacology , Tumor Cells, Cultured
19.
J Biol Chem ; 274(29): 20643-9, 1999 Jul 16.
Article in English | MEDLINE | ID: mdl-10400696

ABSTRACT

The activation of intracellular calcium release and calcium entry across the plasmalemma in response to intracellular application of inositol 2,4,5-trisphosphate and adenophostin A, two metabolically stable agonists for inositol 1,4,5-trisphosphate receptors, was investigated using Xenopus laevis oocytes and confocal imaging. Intracellular injection of inositol 2,4,5-trisphosphate induced a rapidly spreading calcium signal associated with regenerative calcium waves; the calcium signal filled the peripheral regions of the cell in 1-5 min. Injection of high concentrations of adenophostin A (250 nM) similarly induced rapidly spreading calcium signals. Injection of low concentrations of adenophostin A resulted in calcium signals that spread slowly (>1 h). With extremely low concentrations of adenophostin A (approximately 10 pM), stable regions of Ca2+ release were observed that did not expand to peripheral regions. When the adenophostin A-induced calcium signal was restricted to central regions, compartmentalized calcium oscillations were sometimes observed. Restoration of extracellular calcium caused a rise in cytoplasmic calcium restricted to the region of adenophostin A-induced calcium mobilization. The limited diffusion of adenophostin A provides an opportunity to examine calcium signaling processes under spatially restricted conditions and provides insights into mechanisms of intracellular calcium oscillations and capacitative calcium entry.


Subject(s)
Adenosine/analogs & derivatives , Calcium Signaling/drug effects , Adenosine/pharmacology , Animals , Female , Kinetics , Microscopy, Confocal , Microscopy, Fluorescence , Oocytes/drug effects , Oocytes/metabolism , Xenopus laevis
20.
Bioessays ; 21(1): 38-46, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10070252

ABSTRACT

In the phospholipase C signaling system, Ca(2+) is mobilized from intracellular stores by an action of inositol 1,4,5-trisphosphate. The depletion of intracellular calcium stores activates a calcium entry mechanism at the plasma membrane called capacitative calcium entry. The signal for activating the entry is unknown but likely involves either the generation or release, or both, from the endoplasmic reticulum of some diffusible signal. Recent research has focused on mammalian homologues of the Drosophila TRP protein as potential candidates for capacitative calcium entry channels. This review summarizes current knowledge about the nature of capacitative calcium entry signals, as well as the potential role of mammalian TRP proteins as capacitative calcium entry channel molecules.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Animals , Humans , Ion Channel Gating , Ion Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...