201
|
Dolphin AC. L-type calcium channel modulation. ADVANCES IN SECOND MESSENGER AND PHOSPHOPROTEIN RESEARCH 1999; 33:153-77. [PMID: 10218118 DOI: 10.1016/s1040-7952(99)80009-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Affiliation(s)
- A C Dolphin
- Department of Pharmacology, University College of London, England
| |
Collapse
|
202
|
Ho IH, Murrell-Lagnado RD. Molecular determinants for sodium-dependent activation of G protein-gated K+ channels. J Biol Chem 1999; 274:8639-48. [PMID: 10085101 DOI: 10.1074/jbc.274.13.8639] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-gated inwardly rectifying K+ channels (GIRKs) are activated by a direct interaction with Gbetagamma subunits and also by raised internal [Na+]. Both processes require the presence of phosphatidylinositol bisphosphate (PIP2). Here we show that the proximal C-terminal region of GIRK2 mediates the Na+-dependent activation of both the GIRK2 homomeric channels and the GIRK1/GIRK2 heteromeric channels. Within this region, GIRK2 has an aspartate at position 226, whereas GIRK1 has an asparagine at the equivalent position (217). A single point mutation, D226N, in GIRK2, abolished the Na+-dependent activation of both the homomeric and heteromeric channels. Neutralizing a nearby negative charge, E234S had no effect. The reverse mutation in GIRK1, N217D, was sufficient to restore Na+-dependent activation to the GIRK1N217D/GIRK2D226N heteromeric channels. The D226N mutation did not alter either the single channel properties or the ability of these channels to be activated via the m2-muscarinic receptor. PIP2 dramatically increased the open probability of GIRK1/GIRK2 channels in the absence of Na+ or Gbetagamma but did not preclude further activation by Na+, suggesting that Na+ is not acting simply to promote PIP2 binding to GIRKs. We conclude that aspartate 226 in GIRK2 plays a crucial role in Na+-dependent gating of GIRK1/GIRK2 channels.
Collapse
Affiliation(s)
- I H Ho
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QJ, United Kingdom
| | | |
Collapse
|
203
|
Kotlikoff MI, Herrera G, Nelson MT. Calcium permeant ion channels in smooth muscle. Rev Physiol Biochem Pharmacol 1999; 134:147-99. [PMID: 10087909 DOI: 10.1007/3-540-64753-8_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- M I Kotlikoff
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
204
|
Abstract
M2 receptor stimulation results in the gating of nonselective cation channels in several smooth muscle cell types. However the requirement for current activation includes a rise in cytosolic calcium mediated by M3 receptor induced calcium release. This complex signaling system confers substantial complexity on the interpretation of pharmacological experiments. M2 and M3 receptor stimulation has also been linked to the inhibition of potassium channels in smooth muscle. These signaling events are likely to play important roles in excitation/contraction coupling.
Collapse
Affiliation(s)
- M I Kotlikoff
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104-6046, USA
| | | | | |
Collapse
|
205
|
Sun L, Miller RJ. Multiple neuropeptide Y receptors regulate K+ and Ca2+ channels in acutely isolated neurons from the rat arcuate nucleus. J Neurophysiol 1999; 81:1391-403. [PMID: 10085364 DOI: 10.1152/jn.1999.81.3.1391] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We examined the effects of neuropeptide Y (NPY) and related peptides on Ca2+ and K+ currents in acutely isolated neurons from the arcuate nucleus of the rat. NPY analogues that activated all of the known NPY receptors (Y1-Y5), produced voltage-dependent inhibition of Ca2+ currents and activation of inwardly rectifying K+ currents in arcuate neurons. Both of these effects could occur simultaneously in the same cells. In some cells, activation of Y4 NPY receptors also caused oscillations in [Ca2+]i. NPY hyperpolarized arcuate neurons through the activation of a K+ conductance and increased the spike threshold. Molecular biological studies indicated that arcuate neurons possessed all of the previously cloned NPY receptor types (Y1, Y2, Y4, and Y5). Thus activation of multiple types NPY receptors on arcuate neurons can regulate both Ca2+ and K+ conductances leading to a reduction in neuronal excitability and a suppression of neurotransmitter release.
Collapse
Affiliation(s)
- L Sun
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
206
|
Kennedy ME, Nemec J, Corey S, Wickman K, Clapham DE. GIRK4 confers appropriate processing and cell surface localization to G-protein-gated potassium channels. J Biol Chem 1999; 274:2571-82. [PMID: 9891030 DOI: 10.1074/jbc.274.4.2571] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
GIRK1 and GIRK4 subunits combine to form the heterotetrameric acetylcholine-activated potassium current (IKACh) channel in pacemaker cells of the heart. The channel is activated by direct binding of G-protein Gbetagamma subunits. The GIRK1 subunit is atypical in the GIRK family in having a unique ( approximately 125-amino acid) domain in its distal C terminus. GIRK1 cannot form functional channels by itself but must combine with another GIRK family member (GIRK2, GIRK3, or GIRK4), which are themselves capable of forming functional homotetramers. Here we show, using an extracellularly Flag-tagged GIRK1 subunit, that GIRK1 requires association with GIRK4 for cell surface localization. Furthermore, GIRK1 homomultimers reside in core-glycosylated and nonglycosylated states. Coexpression of GIRK4 caused the appearance of the mature glycosylated form of GIRK1. [35S]Methionine pulse-labeling experiments demonstrated that GIRK4 associates with GIRK1 either during or shortly after subunit synthesis. Mutant and chimeric channel subunits were utilized to identify domains responsible for GIRK1 localization. Truncation of the unique C-terminal domain of Delta374-501 resulted in an intracellular GIRK1 subunit that produced normal IKACh-like channels when coexpressed with GIRK4. Chimeras containing the C-terminal domain of GIRK1 from amino acid 194 to 501 were intracellularly localized, whereas chimeras containing the C terminus of GIRK4 localized to the cell surface. Deletion analysis of the GIRK4 C terminus identified a 25-amino acid region required for cell surface targeting of GIRK1/GIRK4 heterotetramers and a 25-amino acid region required for cell surface localization of GIRK4 homotetramers. GIRK1 appeared intracellular in atrial myocytes isolated from GIRK4 knockout mice and was not maturely glycosylated, supporting an essential role for GIRK4 in the processing and cell surface localization of IKACh in vivo.
Collapse
Affiliation(s)
- M E Kennedy
- Howard Hughes Medical Institute, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
207
|
Sorota S, Rybina I, Yamamoto A, Du XY. Isoprenaline can activate the acetylcholine-induced K+ current in canine atrial myocytes via Gs-derived betagamma subunits. J Physiol 1999; 514 ( Pt 2):413-23. [PMID: 9852323 PMCID: PMC2269084 DOI: 10.1111/j.1469-7793.1999.413ae.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
1. G protein betagamma subunits activate the acetylcholine-induced potassium current IK,ACh. There is no evidence of specificity at the level of the betagamma subunits. Therefore all G protein-coupled receptors in atrial myocytes should be able to activate IK,ACh. Paradoxically, it is often stated that isoprenaline does not activate IK,ACh. Rationales to explain this negative result include insufficient concentrations of Gs in the atrium or restricted access of Gs-derived betagamma subunits to the IK,ACh channel. We took advantage of a non-specific increase in Gs that results after infection with adenovirus. 2. Adenoviral infection unmasked a 1 microM isoprenaline-induced IK,ACh which was prevented by propranolol. Isoprenaline occasionally activated IK,ACh in uninfected and freshly dissociated atrial myocytes but the effect was larger and more consistent in infected myocytes. 3. Pertussis toxin pretreatment (100 ng ml-1 overnight) did not block the effect of isoprenaline. The effect of isoprenaline became persistent if cells were pretreated with cholera toxin (200 ng nl-1). 4. Signal transduction events distal to adenylyl cyclase were not involved in isoprenaline-induced IK,ACh. Forskolin (10 microM) did not activate IK,ACh. Inhibition of adenylyl cyclase with cytoplasmic application of 300 microM 2'-deoxyadenosine 3'-monophosphate did not prevent the activation of IK,ACh by isoprenaline. 5. Cytoplasmic application of a betagamma binding peptide derived from the C terminus of beta-adrenergic receptor kinase 1 (50 microM) prevented the effect of isoprenaline on IK,ACh. The peptide did not prevent the stimulation of the L-type calcium current by isoprenaline. 6. The results indicate that beta-adrenoceptors can activate IK,ACh in atrial myocytes through the release of betagamma subunits from Gs.
Collapse
Affiliation(s)
- S Sorota
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | | | | | | |
Collapse
|
208
|
Chapter 18 Effect of Phosphatidylinositol Phosphates on the Gating of G-Protein-Activated K+Channels. CURRENT TOPICS IN MEMBRANES 1999. [DOI: 10.1016/s0070-2161(08)60934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
209
|
Proks P, Gribble FM, Adhikari R, Tucker SJ, Ashcroft FM. Involvement of the N-terminus of Kir6.2 in the inhibition of the KATP channel by ATP. J Physiol 1999; 514 ( Pt 1):19-25. [PMID: 9831713 PMCID: PMC2269058 DOI: 10.1111/j.1469-7793.1999.019af.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
1. ATP-sensitive potassium (KATP) channels are composed of pore-forming Kir6.2 and regulatory SUR subunits. A truncated isoform of Kir6.2, Kir6.2DeltaC26, expresses ATP-sensitive channels in the absence of SUR1, suggesting the ATP-inhibitory site lies on the Kir6. 2 subunit. 2. We examined the effect on the channel ATP sensitivity of mutating the arginine residue at position 50 (R50) in the N-terminus of Kir6.2, by recording macroscopic currents in membrane patches excised from Xenopus oocytes expressing wild-type or mutant Kir6.2DeltaC26. 3. Substitution of R50 by serine, alanine or glycine reduced the Ki for ATP inhibition from 117 microM to 800 microM, 1.1 mM and 3.8 mM, respectively. The single-channel conductance and kinetics were unaffected by any of these mutations. Mutation to glutamate, lysine, asparagine, glutamine or leucine had a smaller effect (Ki, approximately 300-400 microM). The results indicate that the side chain of the arginine residue at position 50 is unlikely to contribute directly to the binding site for ATP, and suggest it may affect ATP inhibition by allosteric interactions. 4. Mutation of the isoleucine residue at position 49 to glycine (I49G) reduced the channel ATP sensitivity, while the mutation of the glutamate residue at position 51 to glycine (E51G) did not. 5. When a mutation in the N-terminus of Kir6.2DeltaC26 that alters ATP sensitivity (R50S; Ki, 800 microM) was combined with one in the C-terminus (E179Q; Ki, 300 microM), the Ki for the apparent ATP sensitivity was increased to 2.8 mM. The Hill coefficient was also increased. This suggests that the N- and C-termini of Kir6.2 may co-operate to influence channel closure by ATP.
Collapse
Affiliation(s)
- P Proks
- University Laboratory of Physiology, Parks Road, Oxford OX1 3PT,, UK
| | | | | | | | | |
Collapse
|
210
|
Reuveny E, Jan LY. Chapter 17 G-Protein Control of G-Protein-Gated Potassium Channels. CURRENT TOPICS IN MEMBRANES 1999. [DOI: 10.1016/s0070-2161(08)60933-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
211
|
McPhee JC, Dang YL, Davidson N, Lester HA. Evidence for a functional interaction between integrins and G protein-activated inward rectifier K+ channels. J Biol Chem 1998; 273:34696-702. [PMID: 9856991 DOI: 10.1074/jbc.273.52.34696] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heteromultimeric G protein-activated inward rectifier K+ (GIRK) channels, abundant in heart and brain, help to determine the cellular membrane potential as well as the frequency and duration of electrical impulses. The sequence arginine-glycine-aspartate (RGD), located extracellularly between the first membrane-spanning region and the pore, is conserved among all identified GIRK subunits but is not found in the extracellular domain of any other cloned K+ channels. Many integrins, which, like channels, are integral membrane proteins, recognize this RGD sequence on other proteins, usually in the extracellular matrix. We therefore asked whether GIRK activity might be regulated by direct interaction with integrin. Here, we present evidence that mutation of the RGD site to RGE, particularly on the GIRK4 subunit, decreases or abolishes GIRK current. Furthermore, wild-type channels can be co-immunoprecipitated with integrin. The total cellular amount of expressed mutant GIRK channel protein is the same as the wild-type protein; however, the amount of mutant channel protein that localizes to the plasma membrane is decreased relative to wild-type, most likely accounting for the diminished GIRK current detected. GIRK channels appear to bind directly to integrin and to require this interaction for proper GIRK channel membrane localization and function.
Collapse
Affiliation(s)
- J C McPhee
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA
| | | | | | | |
Collapse
|
212
|
Schulte U, Hahn H, Wiesinger H, Ruppersberg JP, Fakler B. pH-dependent gating of ROMK (Kir1.1) channels involves conformational changes in both N and C termini. J Biol Chem 1998; 273:34575-9. [PMID: 9852128 DOI: 10.1074/jbc.273.51.34575] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ROMK channels (Kir1.1) are members of the superfamily of inward rectifier potassium channels (Kir) and represent the channels underlying K+ secretion in the kidney. As their native counterparts, Kir1.1 channels are gated by intracellular pH, with acidification leading to channel closure. Although a lysine residue (Lys80) close to the first hydrophobic segment M1 has been identified as the pH sensor, little is known about how opening and closing of the channel is accomplished. Here we investigate the gating process of Kir1.1 channels exploiting their state-dependent modification by water-soluble oxidants and sulfhydryl reagents. Mutagenesis of all intracellular cysteines either alone or in combination revealed two residues targeted by these reagents, one in the N terminus (Cys49) and one in the C terminus (Cys308) of the channel protein. Both sites reacted with the thiol reagents only in the closed state and not in the open state. These results indicate that pH-dependent gating of Kir1.1 channels involves movement of protein domains in both N and C termini of the Kir1.1 protein.
Collapse
Affiliation(s)
- U Schulte
- Department of Physiology II, Ob dem Himmelreich 7, 72074 Tübingen, Germany
| | | | | | | | | |
Collapse
|
213
|
Ruiz-Velasco V, Ikeda SR. Heterologous expression and coupling of G protein-gated inwardly rectifying K+ channels in adult rat sympathetic neurons. J Physiol 1998; 513 ( Pt 3):761-73. [PMID: 9824716 PMCID: PMC2231322 DOI: 10.1111/j.1469-7793.1998.761ba.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/1998] [Accepted: 09/14/1998] [Indexed: 12/31/2022] Open
Abstract
1. G protein-gated inwardly rectifying K+ (GIRK) channels were heterologously expressed in rat superior cervical ganglion (SCG) neurons by intranuclear microinjection. The properties of GIRK channels and their coupling to native receptors were characterized using the whole-cell patch-clamp technique. 2. Following coinjection of either GIRK1-2 or GIRK1-4 cDNA, application of noradrenaline (NA) produced large inwardly rectifying K+ currents. Injection of cDNA encoding individual GIRK subunits produced only small and inconsistent NA-activated inward currents. Current arising from the native expression of GIRK channels in SCG neurons was not observed. 3. NA-mediated activation of GIRK channels was abolished by pertussis toxin (PTX) pretreatment, indicating coupling via G proteins of the Gi/Go subfamily. Conversely, vasoactive intestinal peptide (VIP) activated GIRK channel currents via a cholera toxin-sensitive pathway suggesting coupling through Galphas. Pretreatment of neurons with PTX caused a significant increase in amplitude of the VIP-mediated GIRK channel currents when compared with untreated cells. 4. Application of adenosine, prostaglandin E2 and somatostatin resulted in activation of GIRK channel currents. Activation of m1 muscarinic acetylcholine receptors (i.e. application of oxotremorine M to PTX-treated neurons) failed to elicit overt GIRK channel currents. 5. GIRK channel overexpression decreased basal Ca2+ channel facilitation significantly when compared with uninjected neurons. Furthermore, the NA-mediated inhibition of Ca2+ channels was significantly attenuated. 6. In summary, the ability to heterologously express GIRK channels in adult sympathetic neurons allows the experimental alteration of receptor-G protein-effector stoichiometry. Such studies may increase our understanding of the mechanisms underlying ion channel modulation by G proteins in a neuronal environment.
Collapse
Affiliation(s)
- V Ruiz-Velasco
- Laboratory of Molecular Physiology, Guthrie Research Institute, One Guthrie Square, Sayre, PA 18840,, USA
| | | |
Collapse
|
214
|
Bender K, Wellner-Kienitz MC, Meyer T, Pott L. Activation of muscarinic K+ current by beta-adrenergic receptors in cultured atrial myocytes transfected with beta1 subunit of heterotrimeric G proteins. FEBS Lett 1998; 439:115-20. [PMID: 9849890 DOI: 10.1016/s0014-5793(98)01350-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Muscarinic K+ channels (IK(ACh)) in native atrial myocytes are activated by betagamma subunits of pertussis toxin (Ptx)-sensitive heterotrimeric G proteins coupled to different receptors. betagamma subunits of Ptx-insensitive Gs, coupled to beta-adrenergic receptors, do not activate native IK(ACh). In atrial myocytes from adult rats transfected with rat brain beta1 subunit IK(ACh) can be activated by stimulation of beta-adrenergic receptors using isoprenaline. This effect is insensitive to Ptx. These findings demonstrate for the first time promiscuous (Ptx-insensitive) coupling of Gsbetagamma to GIRK channels in their native environment.
Collapse
Affiliation(s)
- K Bender
- Institut für Physiologie, Abteilung Zelluläre Physiologie, Ruhr-Universität Bochum, Germany
| | | | | | | |
Collapse
|
215
|
Neurotransmitter activation of inwardly rectifying potassium current in dissociated hippocampal CA3 neurons: interactions among multiple receptors. J Neurosci 1998. [PMID: 9763462 DOI: 10.1523/jneurosci.18-20-08153.1998] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We characterized potassium current activated by G-protein-coupled receptors in acutely dissociated hippocampal CA3 neurons. Agonists for serotonin, adenosine, and somatostatin receptors reliably activated a potassium-selective conductance that was inwardly rectifying and that was blocked by 1 mM external Ba2+. The conductance had identical properties to that activated by GABAB receptors in the same cells. In one-half of the CA3 neurons that were tested, the metabotropic glutamate agonist 1S,3R-ACPD also activated inwardly rectifying Ba2+-sensitive potassium current. Activation of the current by serotonin and adenosine agonists occurred with a time constant of 200-700 msec after a lag of 50-100 msec; on removal of agonist the current deactivated with a time constant of 1-2 sec after a lag of 200-400 msec. These kinetics are similar to GABAB-activated current and consistent with a direct action of G-protein on the channels. For somatostatin, both activation and deactivation were approximately fourfold slower, probably limited by agonist binding and unbinding. The half-maximally effective agonist concentrations were approximately 75 nM for somatostatin, approximately 100 nM for serotonin, and approximately 400 nM for 2-chloroadenosine. Dose-response relationships had Hill coefficients of 1.2-1.9, suggesting cooperativity in the receptor-to-channel coupling mechanism. At saturating concentrations of agonists, the combined application of baclofen and either somatostatin, serotonin, or 2-chloroadenosine produced effects that were subadditive and often completely occlusive. However, at subsaturating concentrations the effects of baclofen and 2-chloroadenosine were supra-additive. Thus, low levels of different transmitters can act synergistically in activating inwardly rectifying potassium current.
Collapse
|
216
|
Brandts B, Brandts A, Wellner-Kienitz MC, Zidek W, Schluter H, Pott L. Non-receptor-mediated activation of IK(ATP) and inhibition of IK(ACh) by diadenosine polyphosphates in guinea-pig atrial myocytes. J Physiol 1998; 512 ( Pt 2):407-20. [PMID: 9763631 PMCID: PMC2231209 DOI: 10.1111/j.1469-7793.1998.407be.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
1. The effects of diadenosine polyphosphates (APnA, where n = 4-6) were studied on beating frequency of perfused guinea-pig hearts and on muscarinic K+ current (IK(ACh)) and ATP-regulated K+ current (IK(ATP)) in atrial myocytes from guinea-pig hearts using whole-cell voltage clamp. 2. Bradycardia induced by APnA in perfused hearts was completely inhibited by 8-cyclopentyl- 1,3-dipropylxanthine (CPX, 20 microM), a selective antagonist at A1 adenosine receptors, and was augmented by dipyridamole (Dipy), an inhibitor of cellular adenosine (Ado) uptake. 3. Whereas exposure of atrial myocytes to Ado (100 microM) within about 1 s induced a significant whole-cell IK(ACh), APnA up to 1 mM applied for some tens of seconds failed to activate IK(ACh). If present for periods > 2 min, APnA caused inhibition of agonist-evoked IK(ACh) and activation of a weakly inward rectifying K+ current, which was identified as IK(ATP) by its sensitivity to glibenclamide and its current-voltage curve. 4. The actions of extracellular APnA on IK(ACh) and IK(ATP) were mimicked by intracellular loading of compounds via the patch clamp pipette and by intracellular loading of AMP. 5. The results from isolated myocytes exclude APnA acting as A1 agonists. It is suggested that myocytes can take up APnA, which are degraded to AMP. In the presence of ATP, AMP is converted to ADP, a physiological activator of ATP-regulated K+ channels, by adenylate kinase. A similar mechanism resulting in a reduction of the [GTP]/[GDP] ratio might be responsible for inhibition of IK(ACh). 6. In the perfused heart and other multicellular cardiac preparations the actions of APnA are mediated by Ado via A1 receptors. It is suggested that APnA in multicellular cardiac tissue are hydrolysed by an ectohydrolase to yield AMP which is converted to Ado by ectonucleotidases.
Collapse
Affiliation(s)
- B Brandts
- Institut fur Physiologie, Abteilung Zellulare Physiologie und Pathophysiologie, Ruhr-Universitat Bochum, D-44780 Bochum, Germany
| | | | | | | | | | | |
Collapse
|
217
|
Kohda Y, Ding W, Phan E, Housini I, Wang J, Star RA, Huang CL. Localization of the ROMK potassium channel to the apical membrane of distal nephron in rat kidney. Kidney Int 1998; 54:1214-23. [PMID: 9767537 DOI: 10.1046/j.1523-1755.1998.00120.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The apical potassium (K+) channels mediate K+ recycling in thick ascending limb (TAL) and K+ secretion in cortical collecting duct (CCD). Recently, the cDNAs for a family of renal K+ channels, ROMK1, -2 and -3, were identified. Based on the biophysical properties and mRNA distribution, it is believed that these ROMK cDNAs encode the apical K+ channels of TAL and CCD. However, the information for cellular and subcellular localization of the ROMK proteins in these tubules is still not available. METHODS Paraffin or frozen kidney sections from adult Sprague-Dawley rats were stained by polyclonal antibodies against the N- and C-terminal domain of ROMK. Immunoreactive staining was visualized by color development from horseradish peroxidase reaction. Membrane homogenates from kidney were analyzed by Western blot analysis. RESULTS The polyclonal antibodies against cytoplasmic epitope of ROMK recognized a approximately 42 kD protein in the membrane homogenates from kidney, but not from liver. Staining by immunocytochemistry revealed that ROMK channels were localized to the apical membranes of the distal nephron in cortex and outer medulla, including thick ascending limb and collecting tubule. ROMK staining was absent in glomerulus, proximal tubule and inner medulla. Double staining of the tissue section with both ROMK-specific and H+-ATPase-specific antibodies revealed labeling of ROMK in the principal cells of the collecting tubules. CONCLUSIONS These results further strengthen the idea that ROMK channels play important roles in the recycling of K+ in TAL and the secretion of K+ in CCD.
Collapse
Affiliation(s)
- Y Kohda
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas 75235-8856, USA
| | | | | | | | | | | | | |
Collapse
|
218
|
Wei J, Hodes ME, Piva R, Feng Y, Wang Y, Ghetti B, Dlouhy SR. Characterization of murine Girk2 transcript isoforms: structure and differential expression. Genomics 1998; 51:379-90. [PMID: 9721208 DOI: 10.1006/geno.1998.5369] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A mutation in the G-protein-linked inwardly rectifying K+ channel 2 gene (Girk2) is the cause of the weaver mouse phenotype. We determined that the originally published Girk2 transcript is composed of five exons. The primary coding exon (designated exon 4a in our system) encodes over two-thirds of the protein. Five different full-length Girk2 transcript isoforms (designated Girk2-1, Girk2A-1, Girk2A-2, Girk2B, and Girk2C) originating from different transcriptional start sites and/or alternative splicing were isolated by cDNA RACE. Several of the transcripts were predicted to encode truncated proteins that may lack some of the G-proteincoupling sequence. Northern blotting and in situ hybridization studies with transcript-specific probes indicated that the transcripts were differentially expressed in both normal and weaver mice. All transcripts tested were expressed in the three major targets of action of the weaver mutation: cerebellum, substantia nigra, and testis. Two of the transcripts, Girk2A-1 and Girk2A-2, encode identical proteins and have a distinct pattern of expression in testis, which suggests that they are associated with specific stages of spermatogenesis. An additional transcript, Girk2D, appears to be brain-specific, not polyadenylated, and highly expressed in cerebellar granule cells.
Collapse
Affiliation(s)
- J Wei
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, 46202-5251, USA
| | | | | | | | | | | | | |
Collapse
|
219
|
Ivanova-Nikolova TT, Nikolov EN, Hansen C, Robishaw JD. Muscarinic K+ channel in the heart. Modal regulation by G protein beta gamma subunits. J Gen Physiol 1998; 112:199-210. [PMID: 9689027 PMCID: PMC2525744 DOI: 10.1085/jgp.112.2.199] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/1997] [Accepted: 06/11/1998] [Indexed: 11/20/2022] Open
Abstract
The membrane-delimited activation of muscarinic K+ channels by G protein beta gamma subunits plays a prominent role in the inhibitory synaptic transmission in the heart. These channels are thought to be heterotetramers comprised of two homologous subunits, GIRK1 and CIR, both members of the family of inwardly rectifying K+ channels. Here, we demonstrate that muscarinic K+ channels in neonatal rat atrial myocytes exhibit four distinct gating modes. In intact myocytes, after muscarinic receptor activation, the different gating modes were distinguished by differences in both the frequency of channel opening and the mean open time of the channel, which accounted for a 76-fold increase in channel open probability from mode 1 to mode 4. Because of the tetrameric architecture of the channel, the hypothesis that each of the four gating modes reflects binding of a different number of Gbeta gamma subunits to the channel was tested, using recombinant Gbeta1 gamma5. Gbeta1 gamma5 was able to control the equilibrium between the four gating modes of the channel in a manner consistent with binding of Gbeta gamma to four equivalent and independent sites in the protein complex. Surprisingly, however, Gbeta1 gamma5 lacked the ability to stabilize the long open state of the channel that is responsible for the augmentation of the mean open time in modes 3 and 4 after muscarinic receptor stimulation. The modal regulation of muscarinic K+ channel gating by Gbeta gamma provides the atrial cells with at least two major advantages: the ability to filter out small inputs from multiple membrane receptors and yet the ability to create the gradients of information necessary to control the heart rate with great precision.
Collapse
Affiliation(s)
- T T Ivanova-Nikolova
- Henry Hood MD Research Program, Department of Cellular and Molecular Physiology, Penn State College of Medicine, Danville, Pennsylvania 17822, USA
| | | | | | | |
Collapse
|
220
|
Krapivinsky G, Kennedy ME, Nemec J, Medina I, Krapivinsky L, Clapham DE. Gbeta binding to GIRK4 subunit is critical for G protein-gated K+ channel activation. J Biol Chem 1998; 273:16946-52. [PMID: 9642257 DOI: 10.1074/jbc.273.27.16946] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cardiac G protein-gated K+ channel, IKACh, is directly activated by G protein beta gamma subunits (Gbeta gamma). IKAChis composed of two inward rectifier K+ channel subunits, GIRK1 and GIRK4. Gbeta gamma binds to both GIRK1 and GIRK4 subunits of the heteromultimeric IKACh. Here we delineate the Gbeta gamma binding regions of IKACh by studying direct Gbeta gamma interaction with native purified IKACh, competition of this interaction with peptides derived from GIRK1 or GIRK4 amino acid sequences, mutational analysis of regions implicated in Gbeta gamma binding, and functional expression of mutated subunits in mammalian cells. Only two GIRK4 peptides, containing amino acids 209-225 or 226-245, effectively competed for Gbeta gamma binding. A single point mutation introduced into GIRK4 at position 216 (C216T) dramatically reduced the potency of the peptide in inhibiting Gbeta gamma binding and Gbeta gamma activation of expressed GIRK1/GIRK4(C216T) channels. Conversion of 5 amino acids in GIRK4 (226-245) to the corresponding amino acids found in the G protein-insensitive IRK1 channel, completely abolished peptide inhibition of Gbeta gamma binding to IKACh and Gbeta gamma activation of GIRK1/mutant GIRK4 channels. We conclude from this data that Gbeta gamma binding to GIRK4 is critical for IKACh activation.
Collapse
Affiliation(s)
- G Krapivinsky
- Howard Hughes Medical Institute, Childrens' Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
221
|
Kuriyama H, Kitamura K, Itoh T, Inoue R. Physiological features of visceral smooth muscle cells, with special reference to receptors and ion channels. Physiol Rev 1998; 78:811-920. [PMID: 9674696 DOI: 10.1152/physrev.1998.78.3.811] [Citation(s) in RCA: 176] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Visceral smooth muscle cells (VSMC) play an essential role, through changes in their contraction-relaxation cycle, in the maintenance of homeostasis in biological systems. The features of these cells differ markedly by tissue and by species; moreover, there are often regional differences within a given tissue. The biophysical features used to investigate ion channels in VSMC have progressed from the original extracellular recording methods (large electrode, single or double sucrose gap methods), to the intracellular (microelectrode) recording method, and then to methods for recording from membrane fractions (patch-clamp, including cell-attached patch-clamp, methods). Remarkable advances are now being made thanks to the application of these more modern biophysical procedures and to the development of techniques in molecular biology. Even so, we still have much to learn about the physiological features of these channels and about their contribution to the activity of both cell and tissue. In this review, we take a detailed look at ion channels in VSMC and at receptor-operated ion channels in particular; we look at their interaction with the contraction-relaxation cycle in individual VSMC and especially at the way in which their activity is related to Ca2+ movements and Ca2+ homeostasis in the cell. In sections II and III, we discuss research findings mainly derived from the use of the microelectrode, although we also introduce work done using the patch-clamp procedure. These sections cover work on the electrical activity of VSMC membranes (sect. II) and on neuromuscular transmission (sect. III). In sections IV and V, we discuss work done, using the patch-clamp procedure, on individual ion channels (Na+, Ca2+, K+, and Cl-; sect. IV) and on various types of receptor-operated ion channels (with or without coupled GTP-binding proteins and voltage dependent and independent; sect. V). In sect. VI, we look at work done on the role of Ca2+ in VSMC using the patch-clamp procedure, biochemical procedures, measurements of Ca2+ transients, and Ca2+ sensitivity of contractile proteins of VSMC. We discuss the way in which Ca2+ mobilization occurs after membrane activation (Ca2+ influx and efflux through the surface membrane, Ca2+ release from and uptake into the sarcoplasmic reticulum, and dynamic changes in Ca2+ within the cytosol). In this article, we make only limited reference to vascular smooth muscle research, since we reviewed the features of ion channels in vascular tissues only recently.
Collapse
Affiliation(s)
- H Kuriyama
- Seinan Jogakuin University, Kokura-Kita, Fukuoka, Japan
| | | | | | | |
Collapse
|
222
|
Bockaert J, Pin JP. [Use of a G-protein-coupled receptor to communicate. An evolutionary success]. COMPTES RENDUS DE L'ACADEMIE DES SCIENCES. SERIE III, SCIENCES DE LA VIE 1998; 321:529-51. [PMID: 9769853 DOI: 10.1016/s0764-4469(98)80455-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Among membrane-bound receptors, the seven transmembrane receptors are the most abundant (several thousand, 1% of the genome). They were the most successful during evolution. They are capable of transducing messages as different as photons, organic odorants, nucleotides, nucleosides, peptides, lipids, proteins, etc. They are catalysts of the GDP/GTP nucleotide exchange on heterotrimeric G proteins. They are therefore also called 'G-protein-coupled receptors' (GPCR). G proteins are composed of three subunits, G alpha and two undissociable subunits, G beta gamma. There are at least three families of GPCR showing no sequence similarity. Among G proteins, some have been crystallized (including under the heterotrimeric form) and their structure as well as their activation mechanisms are well known. The structures of GPCR are less known owing to the difficulty in crystallizing membrane-bound proteins. Indirect studies (mutations, 2D crystallization of rhodopsine, molecular modelling, etc.) lead to a useful model of the 'central core' composed of the seven transmembrane domains and of its structural modifications during activation. The intimate contact zones between GPCR and G proteins include, on the GPCR side, domains of intracellular loops and C-terminal, which are specific for each family and on the G protein side, essentially the N- et C-terminal domains plus the alpha 4-beta 6 loop. GPCR can adopt several 'active' conformations some of them being found in mutated receptors responsible for pathologies.
Collapse
Affiliation(s)
- J Bockaert
- CNRS UPR 9023-CCIPE, Montpellier, France.
| | | |
Collapse
|
223
|
Abstract
Activation of different types of G-protein-linked and ionotropic presynaptic receptors has been shown to regulate neurotransmitter release throughout the central and peripheral nervous systems. In the case of G-protein-linked receptors, three major mechanisms have been suggested: (a) inhibition of Ca channels in the nerve terminal; (b) the activation of presynaptic K channels, resulting in a reduction in the effectiveness of the action potential; and (c) direct modulation of one or more components of the neurotransmitter vesicle release apparatus. In the case of ionotropic presynaptic receptors, inhibition of release may be achieved through depolarization of the terminal and inactivation of Na and Ca channels. Activation of presynaptic ionotropic receptors that are appreciably Ca permeable can also enhance the release of transmitters as a result of their ability to raise [Ca]i in the terminal directly. Many transmitters employ several of these mechanisms, thus allowing considerable flexibility in the presynaptic regulation of transmitter release.
Collapse
Affiliation(s)
- R J Miller
- Department of Pharmacological and Physiological Sciences, University of Chicago, Illinois 60637, USA.
| |
Collapse
|
224
|
Ford CE, Skiba NP, Bae H, Daaka Y, Reuveny E, Shekter LR, Rosal R, Weng G, Yang CS, Iyengar R, Miller RJ, Jan LY, Lefkowitz RJ, Hamm HE. Molecular basis for interactions of G protein betagamma subunits with effectors. Science 1998; 280:1271-4. [PMID: 9596582 DOI: 10.1126/science.280.5367.1271] [Citation(s) in RCA: 360] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Both the alpha and betagamma subunits of heterotrimeric guanine nucleotide-binding proteins (G proteins) communicate signals from receptors to effectors. Gbetagamma subunits can regulate a diverse array of effectors, including ion channels and enzymes. Galpha subunits bound to guanine diphosphate (Galpha-GDP) inhibit signal transduction through Gbetagamma subunits, suggesting a common interface on Gbetagamma subunits for Galpha binding and effector interaction. The molecular basis for interaction of Gbetagamma with effectors was characterized by mutational analysis of Gbeta residues that make contact with Galpha-GDP. Analysis of the ability of these mutants to regulate the activity of calcium and potassium channels, adenylyl cyclase 2, phospholipase C-beta2, and beta-adrenergic receptor kinase revealed the Gbeta residues required for activation of each effector and provides evidence for partially overlapping domains on Gbeta for regulation of these effectors. This organization of interaction regions on Gbeta for different effectors and Galpha explains why subunit dissociation is crucial for signal transmission through Gbetagamma subunits.
Collapse
Affiliation(s)
- C E Ford
- Institute for Neuroscience and Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Huang CL, Feng S, Hilgemann DW. Direct activation of inward rectifier potassium channels by PIP2 and its stabilization by Gbetagamma. Nature 1998; 391:803-6. [PMID: 9486652 DOI: 10.1038/35882] [Citation(s) in RCA: 731] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inward rectifier K+ channels, which modulate electrical activity in many cell types, are regulated by protein kinases, guanine-nucleotide-binding proteins (G proteins) and probably actin cytoskeleton. Generation of phosphatidylinositol 4,5-bisphosphate (PIP2) by ATP-dependent lipid kinases is known to activate inward rectifier K+ channels in cardiac membrane patches. Here we report that several cloned inward rectifier K+ channels directly bind PIP2, and that this binding correlates with channel activity. Application of ATP or PIP2 liposomes activates the cloned channels. Stabilized by lipid phosphatase inhibitors, PIP2 antibodies potently inhibit each channel with a unique rate (GIRK1/4 approximately GIRK2 >> IRK1 approximately ROMK. Consistent with the faster dissociation of PIP2 from the GIRK channels, the carboxy terminus of GIRK1 binds 3H-PIP2 liposomes more weakly than does that of IRK1 or ROMK1. Mutation of a conserved arginine to glutamine at position 188 reduces the ability of ROMK1 to bind PIP2 and increases its sensitivity to inhibition by PIP2 antibodies. Interactions between GIRK channels and PIP2 are modulated by the betagamma subunits of the G protein (Gbetagamma). When GIRK1/4 channels are allowed to run down completely, they are not activated by addition of Gbetagamma alone, but application of PIP2 activates them in minutes without Gbetagamma and in just seconds with Gbetagamma. Finally, coexpression of Gbetagamma with GIRK channels slows the inhibition of K+ currents by PIP2 antibodies by more than 10-fold. Thus Gbetagamma activates GIRK channels by stabilizing interactions between PIP2 and the K+ channel.
Collapse
Affiliation(s)
- C L Huang
- Department of Medicine, University of Texas Southwestern Medical Center at Dallas, 75235, USA.
| | | | | |
Collapse
|
226
|
Sui JL, Petit-Jacques J, Logothetis DE. Activation of the atrial KACh channel by the betagamma subunits of G proteins or intracellular Na+ ions depends on the presence of phosphatidylinositol phosphates. Proc Natl Acad Sci U S A 1998; 95:1307-12. [PMID: 9448327 PMCID: PMC18753 DOI: 10.1073/pnas.95.3.1307] [Citation(s) in RCA: 219] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/1997] [Indexed: 02/05/2023] Open
Abstract
The betagamma subunits of GTP-binding proteins (Gbetagamma) activate the muscarinic K+ channel (KACh) in heart by direct binding to both of its component subunits. KACh channels can also be gated by internal Na+ ions. Both activation mechanisms show dependence on hydrolysis of intracellular ATP. We report that phosphatidylinositol 4,5-bisphosphate (PIP2) mimics the ATP effects and that depletion or block of PIP2 retards the stimulatory effects of Gbetagamma subunits or Na+ ions on channel activity, effects that can be reversed by restoring PIP2. Thus, regulation of KACh channel activity may be crucially dependent on PIP2 and phosphatidylinositol signaling. These striking functional results are in agreement with in vitro biochemical studies on the PIP2 requirement for Gbetagamma stimulation of G protein receptor kinase activity, thus implicating phosphatidylinositol phospholipids as a potential control point for Gbetagamma-mediated signal transduction.
Collapse
Affiliation(s)
- J L Sui
- Department of Physiology and Biophysics, Mount Sinai School of Medicine, City University of New York, New York, NY 10029, USA
| | | | | |
Collapse
|
227
|
Affiliation(s)
- H E Hamm
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Institute for Neuroscience, Northwestern University Medical School, Chicago, Illinois 60611, USA.
| |
Collapse
|
228
|
Abstract
Acetylcholine (ACh) released from the stimulated vagus nerve decreases heart rate via modulation of several types of ion channels expressed in cardiac pacemaker cells. Although the muscarinic-gated potassium channel I(KACh) has been implicated in vagally mediated heart rate regulation, questions concerning the extent of its contribution have remained unanswered. To assess the role of I(KACh) in heart rate regulation in vivo, we generated a mouse line deficient in I(KACh) by targeted disruption of the gene coding for GIRK4, one of the channel subunits. We analyzed heart rate and heart rate variability at rest and after pharmacological manipulation in unrestrained conscious mice using electrocardiogram (ECG) telemetry. We found that I(KACh) mediated approximately half of the negative chronotropic effects of vagal stimulation and adenosine on heart rate. In addition, this study indicates that I(KACh) is necessary for the fast fluctuations in heart rate responsible for beat-to-beat control of heart activity, both at rest and after vagal stimulation. Interestingly, noncholinergic systems also appear to modulate heart activity through I(KACh). Thus, I(KACh) is critical for effective heart rate regulation in mice.
Collapse
Affiliation(s)
- K Wickman
- Department of Cardiology, Harvard Medical School, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
229
|
Vivaudou M, Chan KW, Sui JL, Jan LY, Reuveny E, Logothetis DE. Probing the G-protein regulation of GIRK1 and GIRK4, the two subunits of the KACh channel, using functional homomeric mutants. J Biol Chem 1997; 272:31553-60. [PMID: 9395492 DOI: 10.1074/jbc.272.50.31553] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In heart, G-protein-activated channels are complexes of two homologous proteins, GIRK1 and GIRK4. Expression of either protein alone results in barely active or non-active channels, making it difficult to assess the individual contribution of each subunit to the channel complex. The residue Phe137, located within the H5 region of GIRK1, is critical to the synergy between GIRK1 and GIRK4 (Chan, K. W., Sui, J. L., Vivaudou, M., and Logothetis, D. E. (1996) Proc. Natl. Acad. Sci. U. S. A. 93, 14193-14198). By modifying this residue or the matching residue of GIRK4, Ser143, we have been able to generate mutant proteins that produced large inwardly rectifying, G-protein-modulated currents when expressed alone in Xenopus oocytes. The enhanced activity of the heterologous expression of each of two active mutants, GIRK1(F137S) and GIRK4(S143T), was not caused by association with an endogenous oocyte channel subunit, and these mutants did not display apparent differences in the ability to localize to the cell surface compared with their wild-type counterparts. When these functional mutant channels were compared individually with wild-type heteromeric channels, they responded with only small differences to a number of maneuvers involving coexpression with muscarinic receptors, G-protein betagamma subunits, wild-type or mutated G-protein alpha subunits, and active protomers of pertussis toxin. These experiments, which confirmed the crucial, though not exclusive, role of Gbetagamma in regulating channel activity, demonstrated that GIRK1(F137S) and GIRK4(S143T), and by extrapolation their wild-type counterparts, interact in a qualitatively similar way with G-protein subunits. These findings suggest that functionally important sites of interaction with G-proteins are likely to be located within the homologous regions of GIRK1 and GIRK4 rather than within the divergent terminal regions. They also raise the question of the functional advantage of a heteromeric over homomeric design for G-protein-gated channels.
Collapse
Affiliation(s)
- M Vivaudou
- Department of Physiology and Biophysics, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | |
Collapse
|
230
|
Jakob R, Krieglstein J. Influence of flupirtine on a G-protein coupled inwardly rectifying potassium current in hippocampal neurones. Br J Pharmacol 1997; 122:1333-8. [PMID: 9421279 PMCID: PMC1565078 DOI: 10.1038/sj.bjp.0701519] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. Previous studies have shown that flupirtine, a centrally acting, non-opioid analgesic agent, also exhibits neuroprotective activity in focal cerebral ischaemia in mice and reduces apoptosis induced by NMDA, gp 120 of HIV, prior protein fragment or lead acetate as well as necrosis induced by glutamate or NMDA in cell culture. To study the potential mechanism of the neuroprotective action of flupirtine, we investigated whether flupirtine is able to modulate potassium or NMDA-induced currents in rat cultured hippocampal neurones by use of the whole-cell configuration of the patch-clamp technique. 2. We demonstrated that 1 microM flupirtine activated an inwardly rectifying potassium current (K(ir)) in hippocampal neurones (deltaI=-39+/-18 pA at -130 mV; n=10). This effect was dose-dependent (EC50=0.6 microM). The reversal potential for K(ir) was in agreement with the potassium equilibrium potential predicted from the Nernst equation showing that K(ir) was predominantly carried by K+. Furthermore, the induced current was blocked completely by Ba2+ (1 mM), an effect typical for K(ir). 3. The activation of K(ir) by flupirtine was largely prevented by pretreatment of the cells with pertussis toxin (PTX) indicating the involvement of a PTX-sensitive G-protein in the transduction mechanism (deltaI=-3+/-6 pA at -130 mV; n=8). Inclusion of cyclic AMP in the intracellular solution completely abolished the activation of K(ir) (n=7). 4. The selective alpha2-adrenoceptor antagonist SKF-86466 (10 microM), the selective 5-HT1A antagonist NAN 190 as well as the selective GABA(B) antagonist 2-hydroxysaclofen (10 microM) failed to block the flupirtine effect on the inward rectifier. 5. Flupirtine (1 microM) could not change the current induced by 50 microM NMDA. 6. These results show that in cultured hippocampal neurones flupirtine activates an inwardly rectifying potassium current and that a PTX-sensitive G-protein is involved in the transduction mechanism.
Collapse
Affiliation(s)
- R Jakob
- Institut für Pharmakologie und Toxikologie, Philipps-Universität Marburg, Ketzerbach, Germany
| | | |
Collapse
|
231
|
Abstract
The inwardly rectifying K+ channels of the GIRK (Kir3) family, members of the superfamily of inwardly rectifying K+ channels (Kir), are important physiological tools to regulate excitability in heart and brain by neurotransmitters, and the only ion channels conclusively shown to be activated by a direct interaction with heterotrimeric G protein subunits. During the last decade, especially since their cloning in 1993, remarkable progress has been made in understanding the structure, mechanisms of gating, activation by G proteins, and modulation of these channels. However, much of the molecular details of structure and of gating by G protein subunits and other factors, mechanisms of modulation and desensitization, and determinants of specificity of coupling to G proteins, remain unknown. This review summarizes both the recent advances and the unresolved questions now on the agenda in GIRK studies.
Collapse
Affiliation(s)
- N Dascal
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
232
|
Abstract
This lecture is dedicated to Max Delbrück and Seymour Benzer. Max Delbrück was our graduate advisor. He introduced us to a variety of biophysical problems, and taught us ways of thinking about these problems by example. Potassium channels was one of the topics included in his journal club in the early seventies; Max also carefully considered the feasibility of purifying potassium channels then. It was in Seymour Benzer's laboratory that we began to look for Drosophila mutants that affect synaptic transmission at the larval neuromuscular junction. Shaker was the first behavioural mutant we tested that gave a robust phenotype, a phenotype that could be mimicked by treating wild-type preparations with a potassium channel blocker. This mutant fly has led us to our subsequent molecular studies of potassium channels. Since we settled in the University of California, San Francisco, and began to study neural development as well as potassium channels, we have settled into the pattern of each attending meetings and presenting our studies on one of these two areas so as to avoid both being away from home and our children at the same time. In following this pattern, I will be presenting the studies of potassium channels as part of our long-term collaboration. In this talk I will first briefly take you through the path that led us to the molecular studies of potassium channels and then discuss the diversity and modulation of these potassium channels at the molecular and physiological level.
Collapse
Affiliation(s)
- L Y Jan
- Howard Hughes Medical Institute, University of California, San Francisco 94143-0724, USA
| | | |
Collapse
|
233
|
Luchian T, Dascal N, Dessauer C, Platzer D, Davidson N, Lester HA, Schreibmayer W. A C-terminal peptide of the GIRK1 subunit directly blocks the G protein-activated K+ channel (GIRK) expressed in Xenopus oocytes. J Physiol 1997; 505 ( Pt 1):13-22. [PMID: 9409468 PMCID: PMC1160090 DOI: 10.1111/j.1469-7793.1997.013bc.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. In order to find out the functional roles of cytosolic regions of a G protein-activated, inwardly rectifying potassium channel subunit we studied block of GIRK channels, expressed in Xenopus laevis oocytes, by synthetic peptides in isolated inside-out membrane patches. 2. A peptide (DS6) derived from the very end of the C-terminus of GIRK1 reversibly blocked GIRK activity with IC50 values of 7.9 +/- 2.0 or 3.5 +/- 0.5 micrograms ml-1 (corresponding to 3.7 +/- 0.9 or 1.7 +/- 0.2 mumol l-1) for GIRK1/GIRK5 or GIRK1/GIRK4 channels, respectively. 3. Dose dependency studies of GIRK activation by purified beta gamma subunits of the G protein (G beta gamma) showed that DS6 block of GIRK channels is not the result of competition of the peptide with functional GIRK channels for the available G beta gamma. 4. Burst duration of GIRK channels was reduced, whereas long closed times between bursts were markedly increased, accounting for the channel block observed. 5. Block by the DS6 peptide was slightly voltage dependent, being stronger at more negative potentials. 6. These data support the hypothesis that the distal part of the carboxy-terminus of GIRK1 is a part of the intrinsic gate that keeps GIRK channels closed in the absence of G beta gamma.
Collapse
Affiliation(s)
- T Luchian
- Department for Medical Physics and Biophysics, University of Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
234
|
Kovoor A, Nappey V, Kieffer BL, Chavkin C. Mu and delta opioid receptors are differentially desensitized by the coexpression of beta-adrenergic receptor kinase 2 and beta-arrestin 2 in xenopus oocytes. J Biol Chem 1997; 272:27605-11. [PMID: 9346897 DOI: 10.1074/jbc.272.44.27605] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The Xenopus oocyte expression system was used to test the hypothesis that homologous opioid receptor desensitization results from receptor phosphorylation by G protein-coupled receptor kinases. Activation of delta (DOR), mu (MOR) opioid, or beta2-adrenergic receptors increased K+ conductance in oocytes coexpressing the G protein-gated inwardly rectifying K+ channel subunits GIRK1 and GIRK4, and the intrinsic rate of desensitization was small. Coexpression of beta-adrenergic receptor kinase 2 (beta-ARK2) and beta-arrestin 2 (beta-arr2) synergistically produced a rapid desensitization of both DOR and beta2-adrenergic receptor signaling with a t1/2 < 4 min. beta-ARK2 and beta-arr2 more slowly desensitized MOR responses; a similar synergistic effect on MOR required 2-3 h of agonist treatment. DOR mutants lacking serine and threonine residues at the end of the cytoplasmic tail coupled effectively to GIRK channels but were insensitive to beta-ARK2 and beta-arr2. However, a DOR mutant having serine residues mutated to alanine in the third cytoplasmic loop was indistinguishable in coupling and desensitization from the wild type DOR. These studies establish that opioid receptors can be regulated by beta-ARK2 and beta-arr2 and that a portion of the COOH terminus of DOR enhances sensitivity to this modulation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Arrestins/genetics
- Cell Line
- Cloning, Molecular
- Cyclic AMP-Dependent Protein Kinases/genetics
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalins/pharmacology
- G Protein-Coupled Inwardly-Rectifying Potassium Channels
- Molecular Sequence Data
- Oocytes/metabolism
- Organ Culture Techniques
- Potassium Channels/genetics
- Potassium Channels, Inwardly Rectifying
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/drug effects
- Sequence Homology, Amino Acid
- Xenopus
- beta-Adrenergic Receptor Kinases
- beta-Arrestins
Collapse
Affiliation(s)
- A Kovoor
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280, USA
| | | | | | | |
Collapse
|
235
|
Doupnik CA, Davidson N, Lester HA, Kofuji P. RGS proteins reconstitute the rapid gating kinetics of gbetagamma-activated inwardly rectifying K+ channels. Proc Natl Acad Sci U S A 1997; 94:10461-6. [PMID: 9294233 PMCID: PMC23385 DOI: 10.1073/pnas.94.19.10461] [Citation(s) in RCA: 274] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
G protein-gated inward rectifier K+ (GIRK) channels mediate hyperpolarizing postsynaptic potentials in the nervous system and in the heart during activation of Galpha(i/o)-coupled receptors. In neurons and cardiac atrial cells the time course for receptor-mediated GIRK current deactivation is 20-40 times faster than that observed in heterologous systems expressing cloned receptors and GIRK channels, suggesting that an additional component(s) is required to confer the rapid kinetic properties of the native transduction pathway. We report here that heterologous expression of "regulators of G protein signaling" (RGS proteins), along with cloned G protein-coupled receptors and GIRK channels, reconstitutes the temporal properties of the native receptor --> GIRK signal transduction pathway. GIRK current waveforms evoked by agonist activation of muscarinic m2 receptors or serotonin 1A receptors were dramatically accelerated by coexpression of either RGS1, RGS3, or RGS4, but not RGS2. For the brain-expressed RGS4 isoform, neither the current amplitude nor the steady-state agonist dose-response relationship was significantly affected by RGS expression, although the agonist-independent "basal" GIRK current was suppressed by approximately 40%. Because GIRK activation and deactivation kinetics are the limiting rates for the onset and termination of "slow" postsynaptic inhibitory currents in neurons and atrial cells, RGS proteins may play crucial roles in the timing of information transfer within the brain and to peripheral tissues.
Collapse
Affiliation(s)
- C A Doupnik
- Division of Biology 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| | | | | | | |
Collapse
|
236
|
Stevens EB, Woodward R, Ho IH, Murrell-Lagnado R. Identification of regions that regulate the expression and activity of G protein-gated inward rectifier K+ channels in Xenopus oocytes. J Physiol 1997; 503 ( Pt 3):547-62. [PMID: 9379410 PMCID: PMC1159840 DOI: 10.1111/j.1469-7793.1997.547bg.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. The involvement of the cytoplasmic and core regions of K+ channel Kir3.1 and Kir3.2 subunits in determining the cell surface expression and G protein-gated activity of homomeric and heteromeric channel complexes was investigated by heterologous expression of chimeric and wild-type subunits together with the m2 muscarinic receptor in Xenopus oocytes. 2. Co-expression of Kir3.1 and Kir3.2 subunits yielded currents severalfold larger than those elicited by the individual expression of these subunits. Immunofluorescence labelling indicated that Kir3.2 homomeric channels and Kir3.1-Kir3.2 heteromeric channels were expressed at high levels at the cell surface whereas Kir3.1 homomeric complexes were not expressed at the cell surface. Chimeric subunits composed of Kir3.1 and Kir3.2 showed that the presence of either the cytoplasmic tails or the core region of Kir3.1 in all subunits inhibits expression of channels at the plasma membrane. 3. Substituting the cytoplasmic tails of Kir3.1 for the cytoplasmic tails of Kir3.2, generated a chimeric subunit (121) which displayed dramatically increased acetylcholine-induced channel activity compared with the wild-type Kir3.2 homomeric channel. Cell-attached, single-channel recordings revealed that chimera 121 channel openings were longer than Kir3.2 openings. 4. Individually substituting the N- and C-terminal tails of Kir3.1 for those of Kir3.2 showed that the C-terminal tail of Kir3.1 enhanced the activity of heteromeric channels independently of the N-terminal or core regions of this subunit. 5. The chimeric channel, 121, displayed a higher ratio of ACh-induced to basal activity than the Kir3.1-Kir3.2 or Kir3.2 channels. A smaller proportion of chimera 121 channels appear to be activated by the basal turnover of G proteins, implying that they have a lower affinity for G beta gamma. Our results suggest that substituting the Kir3.1 C-terminal tail for the Kir3.2 tail promotes the opening conformational change of the G beta gamma-bound channel. 6. The core and C-terminal regions of Kir3.1 independently conferred time dependence on voltage-dependent activation. The time constant (tau) was between 5 and 10 ms and varied little over the voltage range -60 to -120 mV.
Collapse
Affiliation(s)
- E B Stevens
- Department of Pharmacology, University of Cambridge, UK
| | | | | | | |
Collapse
|
237
|
Takano K, Yasufuku-Takano J, Kozasa T, Nakajima S, Nakajima Y. Different G proteins mediate somatostatin-induced inward rectifier K+ currents in murine brain and endocrine cells. J Physiol 1997; 502 ( Pt 3):559-67. [PMID: 9279808 PMCID: PMC1159528 DOI: 10.1111/j.1469-7793.1997.559bj.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. Types of G proteins (G protein alpha-subunit subtypes) which mediate the activation of inward rectifier K+ currents by somatostatin (somatotrophin release-inhibiting factor, SRIF) were determined in cultured locus coeruleus neurones from newborn rats and in AtT-20 cells (a mouse pituitary cell line). 2. The whole-cell patch clamp technique was used together with injection of antibodies against pertussis toxin (PTX)-sensitive G protein alpha-subunits or with injection of antisense (or sense) oligonucleotides against these G proteins. 3. In locus coeruleus neurones, the SRIF-induced activation of inward rectifier K+ currents was inhibited by anti-G alpha i1/G alpha i2 antibody injection, but not by anti-G alpha i3 or by anti-G alpha o/G alpha i3 antibody injection, suggesting that the SRIF response is mediated through G alpha i1 and/or G alpha i2. 4. The SRIF-induced activation of the inward rectifier was suppressed in locus coeruleus neurones after injection of antisense oligonucleotides against G alpha i2, but not by injection of sense oligonucleotides against G alpha i2. Injection of antisense (or sense) oligonucleotides against G alpha i1, G alpha i3 and G alpha O (common) had no effect. These results suggest that G alpha i2 is involved in this SRIF response. 5. In AtT-20 cells, the SRIF-induced activation of inward rectifier K+ currents was suppressed by injection of anti-G alpha i3 antibody, but not by injection of anti-G alpha i1/G alpha i2 antibody. 6. The above results indicate that Gi mediates the SRIF effects on inward rectifier K+ currents. However, different subtypes of Gi are involved in the brain neurones and in the endocrine cells: Gi2 in locus coeruleus neurones and Gi3 in AtT-20 cells.
Collapse
Affiliation(s)
- K Takano
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine 60612, USA
| | | | | | | | | |
Collapse
|
238
|
Abstract
The heterotrimeric G proteins are extensively involved in the regulation of cells by extracellular signals. The receptors that control them are often the targets of drugs. There are many isoforms of each of the three subunits that make up these proteins. Thus far, genes for at least sixteen alpha subunits, five beta subunits, and eleven gamma subunits have been identified. In addition, some of these proteins have splice variants or are differentially modified. Based upon what is already known, there are well over a thousand possible G protein heterotrimer combinations. The role of subunit diversity in heterotrimer formation and its effect on signaling by G proteins are still not well understood. However, many current lines of research are leading toward an understanding of these roles. The functional significance of subunit heterogeneity is related to the mechanisms used by G proteins to transmit and integrate the many signals coming into cells through this system. Described here are the basic mechanisms by which G proteins integrate cellular responses, the possible role of subunit heterogeneity in these mechanisms, and the evidence for and against their physiological significance. Recent studies suggest the likely possibility that subunit heterogeneity plays an important role in signaling by G proteins. This role has the potential to extend substantially the flexibility of G proteins in mediating cellular responses to extracellular signals. However, the details of this are yet to be worked out, and they are the subject of many different avenues of research.
Collapse
Affiliation(s)
- J D Hildebrandt
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston 29425-2251, U.S.A.
| |
Collapse
|
239
|
Ma JY, Catterall WA, Scheuer T. Persistent sodium currents through brain sodium channels induced by G protein betagamma subunits. Neuron 1997; 19:443-52. [PMID: 9292732 DOI: 10.1016/s0896-6273(00)80952-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Persistent Na+ currents are thought to be important for integration of neuronal responses. Here, we show that betagamma subunits of G proteins can induce persistent Na+ currents. Coexpression of G beta2gamma3, G beta1gamma3, or G beta5gamma3, but not G beta1gamma1 subunits with rat brain type IIA Na+ channel alpha subunits in tsA-201 cells greatly enhances a component of Na+ current with a normal voltage dependence of activation but with dramatically slowed and incomplete inactivation and with steady-state inactivation shifted +37 mV. Synthetic peptides containing the proposed G betagamma-binding motif, Gln-X-X-Glu-Arg, from either adenylyl cyclase 2 or the Na+ channel alpha subunit C-terminal domain reversed the effect of G beta2gamma3 subunits. These results are consistent with direct binding of G betagamma subunits to the C-terminal domain of the Na+ channel, stabilizing a gating mode responsible for slowed and persistent Na+ current. Modulation of Na+ channel gating by G betagamma subunits is expected to have profound effects on neuronal excitability.
Collapse
Affiliation(s)
- J Y Ma
- Department of Pharmacology, University of Washington, Seattle 98195-7280, USA
| | | | | |
Collapse
|
240
|
Chuang H, Jan YN, Jan LY. Regulation of IRK3 inward rectifier K+ channel by m1 acetylcholine receptor and intracellular magnesium. Cell 1997; 89:1121-32. [PMID: 9215634 DOI: 10.1016/s0092-8674(00)80299-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Inward rectifier K+ channels control the cell's membrane potential and neuronal excitability. We report that the IRK3 but not the IRK1 inward rectifier K+ channel activity is inhibited by m1 muscarinic acetylcholine receptor. This m1 modulation cannot be accounted for by protein kinase C, Ca2+, or channel phosphorylation, but can be mimicked by Mg2+. Based on quantitative analyses of IRK3 and two different IRK1 mutant channels bestowed with sensitivity to m1 modulation, we suggest that the resting Mg2+ level causes chronic inhibition of IRK3 channels, and m1 receptor stimulation may lead to an increase of cytoplasmic Mg2+ concentration and further channel inhibition, due to the ability of Mg2+ to lead these channels into a prolonged inactivated state.
Collapse
Affiliation(s)
- H Chuang
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco 94143-0724, USA
| | | | | |
Collapse
|
241
|
Takano K, Yasufuku-Takano J, Teramoto A, Fujita T. Gi3 mediates somatostatin-induced activation of an inwardly rectifying K+ current in human growth hormone-secreting adenoma cells. Endocrinology 1997; 138:2405-9. [PMID: 9165029 DOI: 10.1210/endo.138.6.5185] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
SRIF activates an inwardly rectifying K+ current in human GH-secreting adenoma cells. Activation of this K+ current induces hyperpolarization of the membrane and abolishment of action potential firing. This mechanism is an essential mechanism for SRIF-induced decrease in intracellular Ca2+ concentration and inhibition of GH secretion. The activation of the inwardly rectifying K+ current is mediated by a pertussis toxin-sensitive G protein. In this article, the expression of the pertussis toxin-sensitive G protein alpha-subunits in the human GH-secreting adenoma cells were analyzed by RT-PCR, and the G protein transducing the SRIF-induced activation of this inwardly rectifying K+ current was investigated. RT-PCR of the messenger RNA from two human GH-secreting adenomas revealed that all G alpha(i1), G alpha(i2), G alpha(i3), and G alpha(o) were expressed in these adenomas. Primary cultured cells from these two adenoma cells were investigated under the voltage clamp of the whole-cell mode. Specific antibodies against the carboxyl terminus of G protein alpha-subunits were microinjected into the cells. Microinjection of antibody against the carboxyl terminal sequence of G alpha(i3) attenuated the SRIF-induced activation of the inwardly rectifying K+ current, whereas antibody against the common carboxyl terminal sequence of G alpha(i1) and G alpha(i2) did not. These data indicate that the G protein transducing the SRIF-induced activation of the inwardly rectifying K+ current is Gi3.
Collapse
Affiliation(s)
- K Takano
- Fourth Department of Internal Medicine, University of Tokyo School of Medicine, Bunkyo-ku, Japan
| | | | | | | |
Collapse
|
242
|
Thibault C, Sganga MW, Miles MF. Interaction of phosducin-like protein with G protein betagamma subunits. J Biol Chem 1997; 272:12253-6. [PMID: 9139665 DOI: 10.1074/jbc.272.19.12253] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Phosducin-like protein (PhLP), a widely expressed ethanol-responsive gene (Miles, M. F., Barhite, S., Sganga, M., and Elliott, M. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 10831-10835), is a homologue of phosducin, a known major regulator of Gbetagamma signaling in retina and pineal gland. However, although phosducin has a well characterized role in retinal phototransduction, function of the PhLP remains unclear. In this study we examine the ability of PhLP to bind Gbetagamma dimer in vitro and in vivo. Using PhLP glutathione S-transferase fusion proteins, we show that PhLP directly binds Gbetagamma in vitro. Studies with a series of truncated PhLP fusion proteins indicate independent binding of Gbetagamma to both the amino- and C-terminal halves of PhLP. Protein-protein interactions between Gbetagamma and PhLP are inhibited by the alpha subunit of Go and Gi3, suggesting that PhLP can bind only free Gbetagamma. Finally, we show that PhLP complexes, at least partially, with Gbetagamma in vivo. Following overexpression of epitope-tagged PhLP together with Gbeta1gamma2 proteins in COS-7 cells, a PhLP-Gbetagamma complex is co-immunoprecipitated by monoclonal antibody directed against the epitope tag. Similarly, polyclonal anti-PhLP antibody co-precipitates endogenous PhLP and Gbetagamma proteins from NG108-15 cell lysates. These data are consistent with the hypothesis that PhLP is a widely expressed modulator of Gbetagamma function. Furthermore, because alternate forms of the PhLP transcript are expressed, there may be functional implications for the existence of two Gbetagamma-binding domains on PhLP.
Collapse
Affiliation(s)
- C Thibault
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, San Francisco, California 94110, USA
| | | | | |
Collapse
|
243
|
Sharon D, Vorobiov D, Dascal N. Positive and negative coupling of the metabotropic glutamate receptors to a G protein-activated K+ channel, GIRK, in Xenopus oocytes. J Gen Physiol 1997; 109:477-90. [PMID: 9101406 PMCID: PMC2219433 DOI: 10.1085/jgp.109.4.477] [Citation(s) in RCA: 125] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Metabotropic glutamate receptors (mGluRs) control intracellular signaling cascades through activation of G proteins. The inwardly rectifying K+ channel, GIRK, is activated by the beta gamma subunits of G proteins and is widely expressed in the brain. We investigated whether an interaction between mGluRs and GIRK is possible, using Xenopus oocytes expressing mGluRs and a cardiac/brain subunit of GIRK, GIRK1, with or without another brain subunit, GIRK2. mGluRs known to inhibit adenylyl cyclase (types 2, 3, 4, 6, and 7) activated the GIRK channel. The strongest response was observed with mGluR2; it was inhibited by pertussis toxin (PTX). This is consistent with the activation of GIRK by Gi/Go-coupled receptors. In contrast, mGluR1a and mGluR5 receptors known to activate phospholipase C, presumably via G proteins of the Gq class, inhibited the channel's activity. The inhibition was preceded by an initial weak activation, which was more prominent at higher levels of mGluR1a expression. The inhibition of GIRK activity by mGluR1a was suppressed by a broad-specificity protein kinase inhibitor, staurosporine, and by a specific protein kinase C (PKC) inhibitor, bis-indolylmaleimide, but not by PTX, Ca(2-)chelation, or calphostin C. Thus, mGluR1a inhibits the GIRK channel primarily via a pathway involving activation of a PTX-insensitive G protein and, eventually, of a subtype of PKC, possibly PKC-mu. In contrast, the initial activation of GIRK1 caused by mGluR1a was suppressed by PTX but not by the protein kinase inhibitors. Thus, this activation probably results from a promiscuous coupling of mGluR1a to a Gi/Go protein. The observed modulations may be involved in the mGluRs effects on neuronal excitability in the brain. Inhibition of GIRK by phospholipase C-activating mGluRs bears upon the problem of specificity of G protein (GIRK interaction) helping to explain why receptors coupled to Gq are inefficient in activating GIRK.
Collapse
Affiliation(s)
- D Sharon
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | | | | |
Collapse
|
244
|
Abstract
Recent advances in the study of receptor-regulated ion channels include the cloning of the genes encoding three types of potassium channel that are favorite targets of receptors for transmitters and hormones. Studies of these channels have also provided a strong indication that G-protein betagamma subunits may gate ion channels via direct protein-protein interactions. Similarities between channel regulation by natriuretic peptides and channel regulation by secreted peptide products of the Alzheimer's beta-amyloid precursor protein offer hints for the existence of a receptor for the latter. There are also other novel examples of channel regulation in excitable and nonexcitable cells, including liver cells and blood cells.
Collapse
Affiliation(s)
- L Y Jan
- Howard Hughes Medical Institute, Department of Physiology, Biochemistry, University of California San, Francisco, CA 94143-0724, USA.
| | | |
Collapse
|
245
|
Wickman K, Seldin MF, Gendler SJ, Clapham DE. Partial structure, chromosome localization, and expression of the mouse Girk4 gene. Genomics 1997; 40:395-401. [PMID: 9073506 DOI: 10.1006/geno.1997.4599] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The G protein-gated potassium channel IKACh constitutes part of a signaling pathway that mediates the negative chronotropic and inotropic effects of acetylcholine on cardiac physiology. Similar or identical ion channels regulate the excitability of many neurons in response to neurotransmitters. IKACh is composed of two homologous subunits, GIRK1 and GIRK4. Here we describe a partial genomic structure of the mouse Girk4 gene. Two exons containing the complete protein-coding sequence were identified. Girk4 was mapped to mouse chromosome 9 (13 cM), consistent with the mapping of human GIRK4 to chromosome 11q23-ter. GIRK4 mRNA was found mainly in mouse heart, with trace levels detected in brain, kidney, lung, and spleen. No detectable levels were observed in skeletal muscle, liver, and testis. The onset of GIRK4 mRNA expression in the developing mouse occurs between Embryonic Days 7 and 11, consistent with the appearance and function of the mouse heart.
Collapse
Affiliation(s)
- K Wickman
- Children's Hospital Medical Center, Harvard Medical School, Enders Building, Room 1309, 320 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
246
|
Chan KW, Sui JL, Vivaudou M, Logothetis DE. Specific regions of heteromeric subunits involved in enhancement of G protein-gated K+ channel activity. J Biol Chem 1997; 272:6548-55. [PMID: 9045681 DOI: 10.1074/jbc.272.10.6548] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Heterologous coexpression of recombinant, G protein-gated, inwardly rectifying K+ (GIRK) channel subunits has yielded large currents, severalfold greater than those obtained from expression of the individual subunits. Such current enhancement has been obtained from coexpression of the inactive GIRK1 subunit with the low activity GIRK2-5 subunits in Xenopus oocytes. Using deletion and chimeric constructs, we now report the identification of a C-terminal region unique to GIRK1 and a larger central region of GIRK4 highly homologous to GIRK1, both of which are critical for production of large currents. Chimeras containing these two regions produced homomeric channels, exhibiting currents severalfold greater than those from either wild-type subunit alone. G protein regulation of such chimeric channel currents resembled that of wild-type currents. Green fluorescent protein-tagged channels showed that the amount of chimeric channel expressed on the oocyte cell surface was similar to its wild-type counterpart, suggesting that the enhanced activity was not due to differences in relative levels of expression but rather to the coexistence of the chimeric regions. Single-channel recordings of the active chimeras exhibited patterns of activities with open-time kinetics and conductance characteristics representative of those of GIRK4, indicating that the presence of the GIRK1 C-terminal region caused an increase in the frequency of channel openings without affecting their duration.
Collapse
Affiliation(s)
- K W Chan
- Department of Physiology and Biophysics, Mount Sinai School of Medicine, City University of New York, New York, New York 10029, USA
| | | | | | | |
Collapse
|
247
|
Drake CT, Bausch SB, Milner TA, Chavkin C. GIRK1 immunoreactivity is present predominantly in dendrites, dendritic spines, and somata in the CA1 region of the hippocampus. Proc Natl Acad Sci U S A 1997; 94:1007-12. [PMID: 9023373 PMCID: PMC19630 DOI: 10.1073/pnas.94.3.1007] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/1996] [Accepted: 11/22/1996] [Indexed: 02/03/2023] Open
Abstract
Electron microscopic analysis of the CA1 region of the rat hippocampus revealed that specific immunoreactivity (IR) for a G protein-gated, inwardly rectifying potassium channel (GIRK1) was present exclusively in neurons and predominantly located in spiny dendrites of pyramidal cells. Within stratum lacunosum-moleculare and the superficial stratum radiatum, GIRK1-IR was often present immediately adjacent to asymmetric (excitatory-type) postsynaptic densities in dendritic spines. The subcellular localization of GIRK1-IR in the Golgi apparatus of pyramidal cell somata and in the plasma membrane of dendrites and dendritic spines confirms the hypothesis that GIRK1 is synthesized by pyramidal cells and transported to the more distal dendritic processes. G protein-coupled receptor activation of a dendritic potassium conductance would attenuate the propagation of excitatory synaptic inputs and thereby produce postsynaptic inhibition. Thus, these results show that the GIRK family of channels joins the list of voltage-sensitive channels now known to be expressed in dendritic spines.
Collapse
Affiliation(s)
- C T Drake
- Department of Neurology and Neuroscience, Cornell University Medical College, New York, NY 10021, USA
| | | | | | | |
Collapse
|
248
|
Abstract
The modulation of a constitutively active IRK1-like inwardly rectifying potassium channel, that is endogenously expressed in the RBL-2H3 cell, was studied with the whole-cell patch-clamp technique. Activation of G-proteins by intracellular application of GTP gamma S revealed a dual modulation of the inward rectifier. An initial increase in inward current amplitude was induced by GTP gamma S, followed by a profound inhibition of the current. The stimulation of the inward rectifier by GTP gamma S was abolished by pretreatment with pertussis toxin. The inhibitory phase of the GTP gamma S-induced response was pertussis toxin-insensitive. Stimulation of the m1-muscarinic receptor expressed in the RBL cell after stable transfection, induced an inhibition of the inwardly rectifying currents. Application of protein kinase C activators such as phorbol 12-myristate 13-acetate and phorbol 12,13-dibutyrate, resulted in a strong inhibition of the currents. Application of the cAMP-dependent protein kinase activator 8-bromo cAMP also induced an inhibition of the inward rectifier. It is concluded that the inward rectifier of the RBL-2H3 cell may be inhibited both by activation of protein kinase C and by cAMP-dependent protein kinase. As this type of inward rectifier is widely expressed in the nervous system, these data imply that the channel can be inhibited by receptors that stimulate phospholipase C and/or stimulate adenylyl cyclase, and can be activated by receptors that inhibit adenylyl cyclase activity.
Collapse
Affiliation(s)
- S V Jones
- Department of Psychiatry, University of Vermont College of Medicine, Burlington 05405, USA
| |
Collapse
|
249
|
Ivanova-Nikolova TT, Breitwieser GE. Effector contributions to G beta gamma-mediated signaling as revealed by muscarinic potassium channel gating. J Gen Physiol 1997; 109:245-53. [PMID: 9041452 PMCID: PMC2220061 DOI: 10.1085/jgp.109.2.245] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Receptor-mediated activation of heterotrimeric G proteins leading to dissociation of the G alpha subunit from G beta gamma is a highly conserved signaling strategy used by numerous extracellular stimuli. Although G beta gamma subunits regulate a variety of effectors, including kinases, cyclases, phospholipases, and ion channels (Clapham, D.E., and E.J. Neer. 1993. Nature (Lond.). 365:403-406), few tools exist for probing instantaneous G beta gamma-effector interactions and little is known about the kinetic contributions of effectors to the signaling process. In this study, we used the atrial muscarinic K + channel, which is activated by direct interactions with G beta gamma subunits (Logothetis, D.E., Y. Kurachi J. Galper, E.J. Neer, and D.E. Clap. 1987. Nature (Lond.). 325:321-326; Wickman, K., J. A. Iniguez-Liuhi, P.A. Davenport, R. Taussig, G.B. Krapivinsky, M.E. Linder, A.G. Gilman, and D.E. Clapham. 1994. Nature (Lond.). 366: 654-663; Huang, C.-L., P.A. Slesinger, P.J. Casey, Y.N. Jan, and L.Y. Jan. 1995. Neuron. 15:1133-1143), as a sensitive reporter of the dynamics of G beta gamma-effector interactions. Muscarinic K+ channels exhibit bursting behavior upon G protein activation, shifting between three distinct functional modes, characterized by the frequency of channel openings during individual bursts. Acetylcholine concentration (and by inference, the concentration of activated G beta gamma) controls the fraction of time spent in each mode without changing either the burst duration or channel gating within individual modes. The picture which emerges is of a G beta gamma effector with allosteric regulation and an intrinsic "off" switch which serves to limit its own activation. These two features combine to establish exquisite channel sensitivity to changes in G beta gamma concentration, and may be indicative of the factors regulating other G beta gamma-modulated effectors.
Collapse
Affiliation(s)
- T T Ivanova-Nikolova
- Johns Hopkins University School of Medicine, Department of Physiology, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
250
|
De Waard M, Liu H, Walker D, Scott VE, Gurnett CA, Campbell KP. Direct binding of G-protein betagamma complex to voltage-dependent calcium channels. Nature 1997; 385:446-50. [PMID: 9009193 DOI: 10.1038/385446a0] [Citation(s) in RCA: 364] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Voltage-dependent Ca2+ channels play a central role in controlling neurotransmitter release at the synapse. They can be inhibited by certain G-protein-coupled receptors, acting by a pathway intrinsic to the membrane. Here we show that this inhibition results from a direct interaction between the G-protein betagamma complex and the pore-forming alpha1 subunits of several types of these channels. The interaction is mediated by the cytoplasmic linker connecting the first and second transmembrane repeats. Within this linker, binding occurs both in the alpha1 interaction domain (AID), which also mediates the interaction between the alpha1 and beta subunits of the channel, and in a second downstream sequence. Further analysis of the binding site showed that several amino-terminal residues in the AID are critical for Gbetagamma binding, defining a site distinct from the carboxy-terminal residues shown to be essential for binding the beta-subunit of the Ca2+ channel. Mutation of an arginine residue within the N-terminal motif abolished betagamma binding and rendered the channel refractory to G-protein modulation when expressed in Xenopus oocytes, showing that the interaction is indeed responsible for G-protein-dependent modulation of Ca2+ channel activity.
Collapse
Affiliation(s)
- M De Waard
- Howard Hughes Medical Institute, University of Iowa College of Medicine, Iowa City 52242, USA
| | | | | | | | | | | |
Collapse
|