Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Cell Mol Med ; 14(10): 2337-49, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20807283

ABSTRACT

The process of store-operated Ca(2+) entry (SOCE), whereby Ca(2+) influx across the plasma membrane is activated in response to depletion of intracellular Ca(2+) stores in the endoplasmic reticulum (ER), has been under investigation for greater than 25 years; however, only in the past 5 years have we come to understand this mechanism at the molecular level. A surge of recent experimentation indicates that STIM molecules function as Ca(2+) sensors within the ER that, upon Ca(2+) store depletion, rearrange to sites very near to the plasma membrane. At these plasma membrane-ER junctions, STIM interacts with and activates SOCE channels of the Orai family. The molecular and biophysical data that have led to these findings are discussed in this review, as are several controversies within this rapidly expanding field.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium/metabolism , TRPC Cation Channels/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Drosophila , Drosophila Proteins/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Membrane Proteins/metabolism , Mice , ORAI1 Protein , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2
2.
Nat Cell Biol ; 11(12): 1465-72, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19881501

ABSTRACT

Store-operated Ca(2+) entry (SOCE) and Ca(2+) release-activated Ca(2+) currents (I(crac)) are strongly suppressed during cell division, the only known physiological situation in which Ca(2+) store depletion is uncoupled from the activation of Ca(2+) influx [corrected]. We found that the endoplasmic reticulum (ER) Ca(2+) sensor STIM1 failed to rearrange into near-plasma membrane puncta in mitotic cells, a critical step in the SOCE-activation pathway. We also found that STIM1 from mitotic cells is recognized by the phospho-specific MPM-2 antibody, suggesting that STIM1 is phosphorylated during mitosis. Removal of ten MPM-2 recognition sites by truncation at amino acid 482 abolished MPM-2 recognition of mitotic STIM1, and significantly rescued STIM1 rearrangement and SOCE response in mitosis. We identified Ser 486 and Ser 668 as mitosis-specific phosphorylation sites, and STIM1 containing mutations of these sites to alanine also significantly rescued mitotic SOCE. Therefore, phosphorylation of STIM1 at Ser 486 and Ser 668, and possibly other sites, underlies suppression of SOCE during mitosis.


Subject(s)
Calcium/metabolism , Membrane Proteins/metabolism , Mitosis , Neoplasm Proteins/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , ORAI1 Protein , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Stromal Interaction Molecule 1
3.
J Physiol ; 587(Pt 10): 2275-98, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19332491

ABSTRACT

Recent studies have defined roles for STIM1 and Orai1 as calcium sensor and calcium channel, respectively, for Ca(2+)-release activated Ca(2+) (CRAC) channels, channels underlying store-operated Ca(2+) entry (SOCE). In addition, these proteins have been suggested to function in signalling and constructing other channels with biophysical properties distinct from the CRAC channels. Using the human kidney cell line, HEK293, we examined the hypothesis that STIM1 can interact with and regulate members of a family of non-selective cation channels (TRPC) which have been suggested to also function in SOCE pathways under certain conditions. Our data reveal no role for either STIM1 or Orai1 in signalling of TRPC channels. Specifically, Ca(2+) entry seen after carbachol treatment in cells transiently expressing TRPC1, TRPC3, TRPC5 or TRPC6 was not enhanced by the co-expression of STIM1. Further, knockdown of STIM1 in cells expressing TRPC5 did not reduce TRPC5 activity, in contrast to one published report. We previously reported in stable TRPC7 cells a Ca(2+) entry which was dependent on TRPC7 and appeared store-operated. However, we show here that this TRPC7-mediated entry was also not dependent on either STIM1 or Orai1, as determined by RNA interference (RNAi) and expression of a constitutively active mutant of STIM1. Further, we determined that this entry was not actually store-operated, but instead TRPC7 activity which appears to be regulated by SERCA. Importantly, endogenous TRPC activity was also not regulated by STIM1. In vascular smooth muscle cells, arginine-vasopressin (AVP) activated non-selective cation currents associated with TRPC6 activity were not affected by RNAi knockdown of STIM1, while SOCE was largely inhibited. Finally, disruption of lipid rafts significantly attenuated TRPC3 activity, while having no effect on STIM1 localization or the development of I(CRAC). Also, STIM1 punctae were found to localize in regions distinct from lipid rafts. This suggests that TRPC signalling and STIM1/Orai1 signalling occur in distinct plasma membrane domains. Thus, TRPC channels appear to be activated by mechanisms dependent on phospholipase C which do not involve the Ca(2+) sensor, STIM1.


Subject(s)
Calcium Channels/physiology , Membrane Proteins/physiology , Neoplasm Proteins/physiology , TRPC Cation Channels/physiology , Arginine Vasopressin/pharmacology , Barium/pharmacology , Calcium Signaling/drug effects , Carbachol/pharmacology , Cell Adhesion Molecules/genetics , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chelating Agents/pharmacology , Diglycerides/pharmacology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Gadolinium/pharmacology , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , ORAI1 Protein , RNA, Small Interfering/genetics , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2 , TRPC6 Cation Channel , Thapsigargin/pharmacology , Transfection , beta-Cyclodextrins/pharmacology
4.
Pflugers Arch ; 457(4): 757-69, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18665391

ABSTRACT

The canonical transient receptor potential (TRPC) proteins have been recognized as key players in calcium entry pathways activated through phospholipase-C-coupled receptors. While it is clearly demonstrated that members of the TRPC3/6/7 subfamily are activated by diacylglycerol, the mechanism by which phospholipase C activates members of the TRPC1/4/5 subfamily remains a mystery. In this paper, we provide evidence for both negative and positive modulatory roles for membrane polyphosphoinositides in the regulation of TRPC5 channels. Depletion of polyphosphatidylinositol 4-phosphate and phosphatidylinositol 4,5-bisphosphate (PIP2) through inhibition of phosphatidylinositol 4-kinase activates calcium entry and membrane currents in TRPC5-expressing but not in TRPC3- or TRPC7-expressing cells. Inclusion of polyphosphatidylinositol 4-phosphate or PIP2, but not phosphatidylinositol 3,4,5-trisphosphate, in the patch pipette inhibited TRPC5 currents. Paradoxically, depletion of PIP2 with a directed 5-phosphatase strategy inhibited TRPC5. Furthermore, when the activity of single TRPC5 channels was examined in excised patches, the channels were robustly activated by PIP2. These findings indicate complex functions for regulation of TRPC5 by PIP2, and we propose that membrane polyphosphoinositides may have at least two distinct functions in regulating TRPC5 channel activity.


Subject(s)
Ion Channel Gating/physiology , Phosphatidylinositol 4,5-Diphosphate/metabolism , TRPC Cation Channels/metabolism , Androstadienes/metabolism , Animals , Antibiotics, Antineoplastic/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Carbachol/metabolism , Cell Line , Cholinergic Agonists/metabolism , Chromones/metabolism , Enzyme Inhibitors/metabolism , Humans , Methacholine Chloride/metabolism , Morpholines/metabolism , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/metabolism , Sirolimus/metabolism , TRPC Cation Channels/genetics , Type C Phospholipases/metabolism , Wortmannin
5.
J Biol Chem ; 283(52): 36071-87, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18940791

ABSTRACT

Intracellular glutathione (GSH) depletion is an important hallmark of apoptosis. We have recently shown that GSH depletion by its extrusion regulates apoptosis independently of excessive reactive oxygen species accumulation. However, the mechanisms by which GSH depletion regulates apoptosis are still unclear. Because disruption of intracellular ionic homeostasis, associated with apoptotic volume decrease (AVD), is necessary for the progression of apoptotic cell death, we sought to evaluate the relationship between GSH transport and ionic homeostasis during Fas ligand (FasL)-induced apoptosis in Jurkat cells. GSH depletion in response to FasL was paralleled by distinct degrees of AVD identified by differences in cellular forward scatter and electronic impedance analysis. Inhibition of GSH efflux prevented AVD, K+ loss, and the activation of two distinct ionic conductances, mediated by Kv1.3 and outward rectifying Cl- channels. Reciprocally, stimulation of GSH loss accelerated the loss of K+, AVD, and consequently the progression of the execution phase of apoptosis. Although high extracellular K+ inhibited FasL-induced apoptosis, GSH depletion was largely independent of K+ loss. These results suggest that deregulation of GSH and ionic homeostasis converge in the regulation of apoptosis in lymphoid cells.


Subject(s)
Apoptosis , Glutathione/metabolism , Ions , Lymphocytes/metabolism , Lymphocytes/pathology , Caspase 3/metabolism , Cell Membrane/metabolism , Cell Separation , Fas Ligand Protein , Flow Cytometry , Humans , Jurkat Cells , Lipids/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Potassium/chemistry , Potassium/metabolism , Shaw Potassium Channels/metabolism
6.
Methods ; 46(3): 204-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18929662

ABSTRACT

Activation of surface membrane receptors coupled to phospholipase C results in the generation of cytoplasmic Ca2+ signals comprised of both intracellular Ca2+ release, and enhanced entry of Ca2+ across the plasma membrane. A primary mechanism for this Ca2+ entry process is attributed to store-operated Ca2+ entry, a process that is activated by depletion of Ca2+ ions from an intracellular store by inositol 1,4,5-trisphosphate. Our understanding of the mechanisms underlying both Ca2+ release and store-operated Ca2+ entry have evolved from experimental approaches that include the use of fluorescent Ca2+ indicators and electrophysiological techniques. Pharmacological manipulation of this Ca2+ signaling process has been somewhat limited; but recent identification of key molecular players, STIM and Orai family proteins, has provided new approaches. Here we describe practical methods involving fluorescent Ca2+ indicators and electrophysiological approaches for dissecting the observed intracellular Ca2+ signal to reveal characteristics of store-operated Ca2+ entry, highlighting the advantages, and limitations, of these approaches.


Subject(s)
Calcium/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Fluorescent Dyes/pharmacology , Humans , Receptors, Calcium-Sensing/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors
7.
J Biol Chem ; 283(28): 19265-73, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18487204

ABSTRACT

Store-operated Ca2+ entry (SOCE) is likely the most common mode of regulated influx of Ca2+ into cells. However, only a limited number of pharmacological agents have been shown to modulate this process. 2-Aminoethyldiphenyl borate (2-APB) is a widely used experimental tool that activates and then inhibits SOCE and the underlying calcium release-activated Ca2+ current (I CRAC). The mechanism by which depleted stores activates SOCE involves complex cellular movements of an endoplasmic reticulum Ca2+ sensor, STIM1, which redistributes to puncta near the plasma membrane and, in some manner, activates plasma membrane channels comprising Orai1, -2, and -3 subunits. We show here that 2-APB blocks puncta formation of fluorescently tagged STIM1 in HEK293 cells. Accordingly, 2-APB also inhibited SOCE and I(CRAC)-like currents in cells co-expressing STIM1 with the CRAC channel subunit, Orai1, with similar potency. However, 2-APB inhibited STIM1 puncta formation less well in cells co-expressing Orai1, indicating that the inhibitory effects of 2-APB are not solely dependent upon STIM1 reversal. Further, 2-APB only partially inhibited SOCE and current in cells co-expressing STIM1 and Orai2 and activated sustained currents in HEK293 cells expressing Orai3 and STIM1. Interestingly, the Orai3-dependent currents activated by 2-APB showed large outward currents at potentials greater than +50 mV. Finally, Orai3, and to a lesser extent Orai1, could be directly activated by 2-APB, independently of internal Ca2+ stores and STIM1. These data reveal novel and complex actions of 2-APB effects on SOCE that can be attributed to effects on both STIM1 as well as Orai channel subunits.


Subject(s)
Boron Compounds/pharmacology , Calcium Channel Agonists/pharmacology , Calcium Channels/metabolism , Calcium/metabolism , Membrane Proteins/metabolism , Calcium Channels/genetics , Cell Line , Humans , Membrane Potentials/drug effects , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , ORAI2 Protein , Stromal Interaction Molecule 1
8.
J Cell Sci ; 121(Pt 6): 762-72, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18285445

ABSTRACT

Stim1 responds to depletion of ER Ca2+ stores by rearranging from tubular structures throughout the ER into punctate structures near the plasma membrane, where it activates Orai store-operated Ca2+ entry (SOCE) channels. However, the mechanism and structural determinants of the localization and reversal of Stim1 puncta formation are poorly understood. Using HEK293 cells expressing Stim1 tagged with enhanced yellow fluorescent protein (EYFP-Stim1), we show that the basis for SOCE termination is the reversal of the punctate Stim1 localization, which absolutely depends on SOCE-dependent store refilling. We also describe rapid, store-independent reversal of EYFP-Stim1 punctae by the ML-9 inhibitor of myosin-light-chain kinase (MLCK). ML-9 similarly inhibited SOCE and the Ca2+-release-activated Ca2+ (CRAC) current. Reversal by ML-9 resulted in full re-establishment of the tubular EYFP-Stim1 localization. A constitutively active EF-hand mutant of EYFP-Stim1 was also reversed by ML-9, regardless of the Ca2+ store content. Inhibition by ML-9 was not due to MLCK inhibition as other inhibitors of MLCK had no effect. Finally, we provide evidence that EYFP-Stim1 punctae form in specific predetermined cellular loci. We conclude that SOCE is tightly coupled to formation of Stim1 puncta, and both SOCE and puncta formation involve a dynamic, reversible signaling complex that probably consists of components in addition to Stim1 and Orai channels.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Membrane Proteins/analysis , Membrane Proteins/metabolism , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Azepines/pharmacology , Bacterial Proteins/analysis , Biological Transport/drug effects , Cell Line , Electric Conductivity , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Humans , Luminescent Proteins/analysis , Membrane Proteins/antagonists & inhibitors , Mutation , Myosin-Light-Chain Kinase/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Patch-Clamp Techniques , Stromal Interaction Molecule 1
9.
Nat Immunol ; 9(1): 89-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18059270

ABSTRACT

CRACM1 (also called Orai1) constitutes the pore subunit of store-operated calcium release-activated calcium channels. A point mutation in the gene encoding CRACM1 is associated with severe combined immunodeficiency disease in humans. Here we generated CRACM1-deficient mice in which beta-galactosidase activity 'reported' CRACM1 expression. CRACM1-deficient mice were smaller in size. Mast cells derived from CRACM1-deficient mice showed grossly defective degranulation and cytokine secretion, and the allergic reactions elicited in vivo were inhibited in CRACM1-deficient mice. We detected robust CRACM1 expression in skeletal muscles and some regions of the brain, heart and kidney but not in the lymphoid regions of thymus and spleen. In contrast, we found CRACM2 expression to be much higher in mouse T cells. In agreement with those findings, the store-operated calcium influx and development and proliferation of CRACM1-deficient T cells was unaffected. Thus, CRACM1 is crucial in mouse mast cell effector function, but mouse T cell calcium release-activated calcium channels are functional in the absence of CRACM1.


Subject(s)
Calcium Channels/physiology , Mast Cells/immunology , Animals , Calcium/metabolism , Calcium Channels/biosynthesis , Cell Degranulation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Mast Cells/metabolism , Mice , Mice, Knockout , ORAI1 Protein , ORAI2 Protein , Organ Specificity , Protein Subunits/biosynthesis , Protein Subunits/physiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
10.
J Cell Sci ; 120(Pt 21): 3762-71, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17925382

ABSTRACT

We examined the role of the microtubule cytoskeleton in the localization and store-operated Ca(2+) entry (SOCE) function of the endoplasmic reticulum (ER) Ca(2+) sensor stromal interaction molecule 1 (STIM1) in HEK 293 cells. STIM1 tagged with an enhanced yellow fluorescent protein (EYFP-STIM1) exhibited a fibrillar localization that colocalized with endogenous alpha-tubulin. Depolymerization of microtubules with nocodazole caused a change from a fibrillar EYFP-STIM1 localization to one that was similar to that of the ER. Treatment of HEK 293 cells with nocodazole had a detrimental impact on SOCE and the associated Ca(2+) release-activated Ca(2+) current (I(CRAC)). This inhibition was significantly reversed in cells overexpressing EYFP-STIM1, implying that the primary inhibitory effect of nocodazole is related to STIM1 function. Surprisingly, nocodazole treatment alone induced significant SOCE and I(CRAC) in cells expressing EYFP-STIM1, and this was accompanied by an increase in EYFP-STIM1 fluorescence near the plasma membrane. We conclude that microtubules play a facilitative role in the SOCE signaling pathway by optimizing the localization of STIM1.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Cytoskeleton/metabolism , Membrane Proteins/metabolism , Microtubules/metabolism , Neoplasm Proteins/metabolism , Calcium/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Nocodazole/metabolism , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stromal Interaction Molecule 1 , Tubulin/metabolism , Tubulin Modulators/metabolism
11.
J Biol Chem ; 282(24): 17548-56, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17452328

ABSTRACT

The recent discoveries of Stim1 and Orai proteins have shed light on the molecular makeup of both the endoplasmic reticulum Ca(2+) sensor and the calcium release-activated calcium (CRAC) channel, respectively. In this study, we investigated the regulation of CRAC channel function by extracellular Ca(2+) for channels composed primarily of Orai1, Orai2, and Orai3, by co-expressing these proteins together with Stim1, as well as the endogenous channels in HEK293 cells. As reported previously, Orai1 or Orai2 resulted in a substantial increase in CRAC current (I(crac)), but Orai3 failed to produce any detectable Ca(2+)-selective currents. However, sodium currents measured in the Orai3-expressing HEK293 cells were significantly larger in current density than Stim1-expressing cells. Moreover, upon switching to divalent free external solutions, Orai3 currents were considerably more stable than Orai1 or Orai2, indicating that Orai3 channels undergo a lesser degree of depotentiation. Additionally, the difference between depotentiation from Ca(2+) and Ba(2+) or Mg(2+) solutions was significantly less for Orai3 than for Orai1 or -2. Nonetheless, the Na(+) currents through Orai1, Orai2, and Orai3, as well as the endogenous store-operated Na(+) currents in HEK293 cells, were all inhibited by extracellular Ca(2+) with a half-maximal concentration of approximately 20 mum. We conclude that Orai1, -2, and -3 channels are similarly inhibited by extracellular Ca(2+), indicating similar affinities for Ca(2+) within the selectivity filter. Orai3 channels appeared to differ from Orai1 and -2 in being somewhat resistant to the process of Ca(2+) depotentiation.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Calcium Channels/genetics , Cations, Divalent/metabolism , Cell Line , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , ORAI1 Protein , ORAI2 Protein , Patch-Clamp Techniques , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stromal Interaction Molecule 1
12.
Biochim Biophys Acta ; 1763(11): 1147-60, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17034882

ABSTRACT

Depletion of intracellular Ca2+ stores induces Ca2+ influx across the plasma membrane through store-operated channels (SOCs). This store-operated Ca2+ influx is important for the replenishment of the Ca2+ stores, and is also involved in many signaling processes by virtue of the ability of intracellular Ca2+ to act as a second messenger. For many years, the molecular identities of particular SOCs, as well as the signaling mechanisms by which these channels are activated, have been elusive. Recently, however, the mammalian proteins STIM1 and Orai1 were shown to be necessary for the activation of store-operated Ca2+ entry in a variety of mammalian cells. Here we present molecular, pharmacological, and electrophysiological properties of SOCs, with particular focus on the roles that STIM1 and Orai1 may play in the signaling processes that regulate various pathways of store-operated entry.


Subject(s)
Calcium Channels/physiology , Calcium Signaling , Calcium/metabolism , Channelopathies/etiology , Membrane Proteins/physiology , Transient Receptor Potential Channels/physiology , Animals , Cell Membrane/metabolism , Humans , Membrane Proteins/genetics
13.
J Biol Chem ; 281(34): 24979-90, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16807233

ABSTRACT

The molecular nature of store-operated Ca(2+)-selective channels has remained an enigma, due largely to the continued inability to convincingly demonstrate Ca(2+)-selective store-operated currents resulting from exogenous expression of known genes. Recent findings have implicated two proteins, Stim1 and Orai1, as having essential roles in store-operated Ca(2+) entry across the plasma membrane. However, transient overexpression of these proteins on their own results in little or no increase in store-operated entry. Here we demonstrate dramatic synergism between these two mediators; co-transfection of HEK293 cells with Stim1 and Orai1 results in an approximate 20-fold increase in store-operated Ca(2+) entry and Ca(2+)-selective current. This demonstrates that these two proteins are limiting for both the signaling and permeation mechanisms for Ca(2+)-selective store-operated Ca(2+) entry. There are three mammalian homologs of Orai1, and in expression experiments they all produced or augmented store-operated Ca(2+) entry with efficacies in the order Orai1 > Orai2 > Orai3. Stim1 apparently initiates the signaling process by acting as a Ca(2+) sensor in the endoplasmic reticulum. This results in rearrangement of Stim1 within the cell and migration toward the plasma membrane to regulate in some manner Orai1 located in the plasma membrane. However, we demonstrate that Stim1 does not incorporate in the surface membrane, and thus likely regulates or interacts with Orai1 at sites of close apposition between the plasma membrane and an intracellular Stim1-containing organelle.


Subject(s)
Calcium Signaling , Calcium/metabolism , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Biological Transport , Calcium Channels , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Humans , ORAI1 Protein , ORAI2 Protein , Stromal Interaction Molecule 1
14.
Neuropeptides ; 38(5): 289-97, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15464194

ABSTRACT

The potency and mechanism of action of vasoactive intestinal peptide (VIP) for producing coronary vasodilation was investigated in the isolated perfused heart of the rat. VIP reduced coronary vascular resistance in a dose-dependent manner, starting at 1 x 10(-10) M, and maximally reduced coronary vascular resistance by 49% at 1 x 10(-8) M. The potency of VIP for reducing coronary vascular resistance (EC50=3.02 x 10(-10) M) was considerably greater than that of adenosine (EC50=6.17 x 10(-8) M) and sodium nitroprusside (EC50=2.45 x 10(-6) M). The vasodilatory action of VIP was more easily observed after increasing vascular tone by perfusion of the hearts with a modified physiological solution containing reduced concentrations of potassium (3.2 mM) and calcium (1.2 mM). Under these conditions, VIP maximally reduced coronary resistance by 54% at 7 x 10(-9) M. The potency of VIP for reducing coronary resistance in these hearts, however, decreased 16-fold (EC50=4.90 x 10(-9) M) while that of SNP remained unaltered (EC50=3.39 x 10(-6) M), compared with hearts perfused with higher levels of potassium (5.9 mM) and calcium (2.5 mM). The vasodilatory effect of VIP occurred without a significant change in heart rate, myocardial contractility or oxygen consumption. In additional studies, the dose-dependent effect of VIP on cyclic nucleotide release from the heart was determined by infusing VIP into the coronary circulation in a cumulative fashion to produce final concentrations between 1 x 10(-11) and 1 x 10(-9) M. VIP increased cyclic AMP at 1 x 10(-9) M but did not increase cyclic GMP. Studies using RT-PCR and immunohistochemistry clearly demonstrated the presence of two VIP receptor subtypes, VPAC1 and VPAC2, in the arteries and arterioles of the heart. In conclusion, VIP is a potent vasodilator in the coronary circulation of the rat and the role of VIP in the control of coronary vascular resistance depends on the circulating levels of potassium and calcium. This vasodilatory effect involves binding to specific coronary cell surface receptors, VPAC1 and/or VPAC2, and is dependent on cyclic AMP only during maximal vasodilation.


Subject(s)
Coronary Circulation/drug effects , Heart/drug effects , Myocardium/metabolism , Vasoactive Intestinal Peptide/pharmacology , Vasodilator Agents/pharmacology , Adenosine/pharmacology , Animals , Calcium/metabolism , Coronary Vessels/cytology , Coronary Vessels/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Nitroprusside/pharmacology , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I
15.
J Biol Chem ; 279(39): 40609-21, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15280371

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) have been found within mammalian intracardiac ganglia, but the cellular effects of these neuropeptides remain poorly understood. Fluorometric calcium imaging and whole cell patch clamp recordings were used to examine the effects of PACAP and VIP on [Ca2+]i and neuroexcitability, respectively, in intracardiac neurons of neonatal rats. PACAP and VIP evoked rapid increases in [Ca2+]i that exhibited both transient and sustained components. Pharmacological experiments using PAC1 and VPAC receptor-selective antagonists demonstrated that the elevations in [Ca2+]i result from the activation of VPAC receptors. The transient increases in [Ca2+]i were shown to be the product of Ca2+ mobilization from caffeine/ryanodine-sensitive intracellular stores and were not due to inositol 1,4,5-trisphosphate-mediated calcium release. In contrast, the sustained [Ca2+]i elevations were dependent on extracellular Ca2+ and were blocked by the transient receptor channel antagonist, 2-aminoethoxydiphenyl borate, which suggests that they are due to Ca2+ entry via store-operated channels. In addition to elevating [Ca2+]i, both PACAP and VIP depolarized intracardiac neurons, and PACAP was further shown to augment action potential firing in these cells. Depolarization of intracardiac neurons by the neuropeptides was dependent on activation of VPAC receptors and the concomitant increases in [Ca2+]i. Although activation of PAC1 receptors alone had no direct effects on neuroexcitability, PAC1 receptor stimulation potentiated the VPAC receptor-induced depolarizations. Furthermore, enhanced action potential firing was only observed upon concurrent stimulation of PAC1 and VPAC receptors, which indicates that these receptors act synergistically to enhance neuroexcitability in intracardiac neurons.


Subject(s)
Heart/innervation , Neurons/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Animals , Animals, Newborn , Boron Compounds/pharmacology , Caffeine/pharmacology , Calcium/chemistry , Calcium/metabolism , Cell Membrane/metabolism , Central Nervous System Stimulants/pharmacology , Electrophysiology , Fluorometry , Membrane Potentials , Peptides/chemistry , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Polypeptide, Type I , Ryanodine/pharmacology , Time Factors
16.
Neurosci Lett ; 328(1): 45-9, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12123856

ABSTRACT

The expression of receptors for pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) was investigated in isolated parasympathetic neurons of neonatal rat intracardiac ganglia using single-cell reverse transcription-polymerase chain reaction. Individual neurons were shown to express multiple isoforms of the PACAP receptor, PAC1, including PAC1-short, -HOP1 and -HOP2 variants, which differ in the region encoding the G protein-binding domain. The PAC1-HOP1 isoform was the predominant species, being expressed at higher levels and in a greater number of cells than other PAC1 variants. In addition to PAC1, intrinsic cardiac neurons express transcripts for the VIP receptors, VPAC1 and VPAC2, with VPAC2 being found in a greater proportion of the neurons. These findings may explain the complex effects of PACAP and VIP on neuroexcitability in mammalian intracardiac ganglia.


Subject(s)
Ganglia, Parasympathetic/metabolism , Heart/innervation , Neurons/metabolism , Neuropeptides/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism , Alternative Splicing/genetics , Animals , Animals, Newborn , Ganglia, Parasympathetic/cytology , Gene Expression Regulation/physiology , Heart/physiology , Neurons/cytology , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Isoforms/genetics , RNA, Messenger/metabolism , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL