1
|
Chen X, Bründl M, Friesacher T, Stary-Weinzinger A. Computational Insights Into Voltage Dependence of Polyamine Block in a Strong Inwardly Rectifying K + Channel. Front Pharmacol 2020; 11:721. [PMID: 32499707 PMCID: PMC7243266 DOI: 10.3389/fphar.2020.00721] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/30/2020] [Indexed: 12/30/2022] Open
Abstract
Inwardly rectifying potassium (KIR) channels play important roles in controlling cellular excitability and K+ ion homeostasis. Under physiological conditions, KIR channels allow large K+ influx at potentials negative to the equilibrium potential of K+ but permit little outward current at potentials positive to the equilibrium potential of K+, due to voltage dependent block of outward K+ flux by cytoplasmic polyamines. These polycationic molecules enter the KIR channel pore from the intracellular side. They block K+ ion movement through the channel at depolarized potentials, thereby ensuring, for instance, the long plateau phase of the cardiac action potential. Key questions concerning how deeply these charged molecules migrate into the pore and how the steep voltage dependence arises remain unclear. Recent MD simulations on GIRK2 (=Kir3.2) crystal structures have provided unprecedented details concerning the conduction mechanism of a KIR channel. Here, we use MD simulations with applied field to provide detailed insights into voltage dependent block of putrescine, using the conductive state of the strong inwardly rectifying K+ channel GIRK2 as starting point. Our µs long simulations elucidate details about binding sites of putrescine in the pore and suggest that voltage-dependent rectification arises from a dual mechanism.
Collapse
|
2
|
Kuzmin VS, Egorov YV, Rozenshtraukh LV. [Electrhopysiological Effect of the Polyamine Spermine in Normoxic and Ischemic Ventricular Myocardium]. ACTA ACUST UNITED AC 2019; 59:43-51. [PMID: 30990140 DOI: 10.18087/cardio.2019.3.10240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 04/13/2019] [Indexed: 11/18/2022]
Abstract
Cytoplasmic polyamines (PA) are involved in control of many cellular functions and are well known as regulators of so called inward-rectifier potassium ion channels. Nevertheless, functional significance of extracellular PA in the heart is poorly elucidated. Aim of this study was to study effects of endogenous PA spermine in the ventricular myocardium. Effects of the extracellular spermine were investigated in isolated multicellular preparations of rabbit and rat ventricular myocardium. Langendorff-perfused isolated rat and rabbit hearts were also used. Action potential (APs) duration and pattern of excitation in ventricular myocardium were estimated using standard microelectrode technique and optical mapping. Functional refractory periods were assessed in Langendorff perfused hearts with the help of programmedelectrical stimulation of the ventricle. In this study extracellular PA spermine (0.1-5 mM) induced shortening of the APs in multicellular preparations of rat ventricular myocardium registered using sharp microelectrode technique. However, spermine caused only weak effect in preparations of ventricular myocardium from rabbit heart: highest tested concentration of spermine (5 mM) induced 4.7 % APs shortening. Similarly, 0.1-1 mM of spermine was unable to alter substantially ventricular effective refractory periods in isolated perfused rabbit hearts. In two animal species tested (rat and rabbit) 0.1-1 mM of spermine failed to affect conduction velocity and activation pattern in ventricles of isolated Langendorff-perfused hearts under normoxia. However, in the rat no-flow model of ischemia-reperfusion extracellular spermine improved conduction of excitation in ventricles. Our results allow suggesting that extracellular spermine can prevent ischemia-induced proarrhythmic changes in ventricular myocardium probably due to reduction of calcium accumulation, but this effect is significant only when PA is applied in millimolar concentrations. Also, potential anti-ischemic effect of the PA may be species specific.
Collapse
Affiliation(s)
| | - Yu V Egorov
- Institute of Experimental Cardiology of National Medical Research Center for Cardiology
| | - L V Rozenshtraukh
- Institute of Experimental Cardiology of National Medical Research Center for Cardiology
| |
Collapse
|
3
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
4
|
Vanheiden S, Pott L, Kienitz MC. Voltage-dependent open-channel block of G protein-gated inward-rectifying K(+) (GIRK) current in rat atrial myocytes by tamoxifen. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:1149-60. [PMID: 23096593 DOI: 10.1007/s00210-012-0801-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/09/2012] [Indexed: 01/15/2023]
Abstract
Tamoxifen (Tmx) is a nonsteroidal selective estrogen receptor antagonist and is frequently used in the treatment and prevention of breast cancer. The compound and its metabolites have been reported to inhibit functions of different classes of membrane proteins, including various ion channels. For members of the inward-rectifying K(+) (Kir) channel family, interference of Tmx with binding of phosphatidylinositol 4,5-bisphosphate (PIP(2)) has been suggested as the mechanism underlying such inhibition. We have studied the inhibition of G protein-activated K(+) (GIRK) current by Tmx in isolated myocytes from hearts of adult rats using whole-cell voltage clamp and experimental conditions for measuring K(+) currents as inward currents (E (K) -50 mV; holding potential -90 mV). Extracellular Tmx reversibly inhibited GIRK current activated by acetylcholine (I (K(ACh))) with an EC(50) of 7.4 × 10(-7) M. This inhibition was composed of two components, a basal reduction in peak current and a block that required opening of channels by ACh. The open-channel block was partially relieved by depolarizing voltage steps in a voltage- and time-dependent fashion. A voltage-dependent open-channel block was not observed when I (K(ACh)) was measured as outward current (E (K) -90 mV; holding potential -40 mV). Intracellular application of Tmx via the patch clamp pipette at a concentration (7 × 10(-6) M) that caused a rapid inhibition of I (K(ACh)) upon extracellular application did not affect the current. Intracellular application of the H(2)O-soluble PIP(2) analog diC(8)-PIP(2) reduced the voltage-independent component of inhibition but had no effect on voltage-dependent open-channel block. The effects of 4-hydroxy-Tmx, a major active metabolite, tested at 2 × 10(-6) M, had effects on I (K(ACh)) analogous to those of Tmx. Inhibition of constitutive inward-rectifying K(+) current (I (K1)) in ventricular myocytes, carried by Kir2 complexes, by Tmx was devoid of a voltage-dependent component. This study suggests the voltage-dependent open-channel block of GIRK inward current as a novel mechanism of Tmx action.
Collapse
Affiliation(s)
- Svenja Vanheiden
- Institute of Physiology, Ruhr-University Bochum, 44780, Bochum, Germany
| | | | | |
Collapse
|
5
|
Hertel F, Switalski A, Mintert-Jancke E, Karavassilidou K, Bender K, Pott L, Kienitz MC. A genetically encoded tool kit for manipulating and monitoring membrane phosphatidylinositol 4,5-bisphosphate in intact cells. PLoS One 2011; 6:e20855. [PMID: 21695261 PMCID: PMC3111442 DOI: 10.1371/journal.pone.0020855] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 05/10/2011] [Indexed: 12/20/2022] Open
Abstract
Background Most ion channels are regulated by phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) in the cell membrane by diverse mechanisms. Important molecular tools to study ion channel regulation by PtdIns(4,5)P2 in living cells have been developed in the past. These include fluorescent PH-domains as sensors for Förster resonance energy transfer (FRET), to monitor changes in plasma membrane. For controlled and reversible depletion of PtdIns(4,5)P2, voltage-sensing phosphoinositide phosphatases (VSD) have been demonstrated as a superior tool, since they are independent of cellular signaling pathways. Combining these methods in intact cells requires multiple transfections. We used self-cleaving viral 2A-peptide sequences for adenovirus driven expression of the PH-domain of phospholipase-Cδ1 (PLCδ1) fused to ECFP and EYFP respectively and Ciona intestinalis VSP (Ci-VSP), from a single open reading frame (ORF) in adult rat cardiac myocytes. Methods and Results Expression and correct targeting of ECFP-PH-PLCδ1, EYFP-PH-PLCδ1, and Ci-VSP from a single tricistronic vector containing 2A-peptide sequences first was demonstrated in HEK293 cells by voltage-controlled FRET measurements and Western blotting. Adult rat cardiac myocytes expressed Ci-VSP and the two fluorescent PH-domains within 4 days after gene transfer using the vector integrated into an adenoviral construct. Activation of Ci-VSP by depolarization resulted in rapid changes in FRET ratio indicating depletion of PtdIns(4,5)P2 in the plasma membrane. This was paralleled by inhibition of endogenous G protein activated K+ (GIRK) current. By comparing changes in FRET and current, a component of GIRK inhibition by adrenergic receptors unrelated to depletion of PtdIns(4,5)P2 was identified. Conclusions Expression of a FRET sensor pair and Ci-VSP from a single ORF provides a useful approach to study regulation of ion channels by phosphoinositides in cell lines and transfection-resistant postmitotic cells. Generally, adenoviral constructs containing self-cleaving 2A-peptide sequences are highly suited for simultaneous transfer of multiple genes in adult cardiac myocytes.
Collapse
Affiliation(s)
- Fabian Hertel
- Institute of Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Agathe Switalski
- Institute of Physiology, Ruhr-University Bochum, Bochum, Germany
| | | | | | - Kirsten Bender
- Institute of Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Lutz Pott
- Institute of Physiology, Ruhr-University Bochum, Bochum, Germany
- * E-mail:
| | | |
Collapse
|
6
|
Hibino H, Inanobe A, Furutani K, Murakami S, Findlay I, Kurachi Y. Inwardly rectifying potassium channels: their structure, function, and physiological roles. Physiol Rev 2010; 90:291-366. [PMID: 20086079 DOI: 10.1152/physrev.00021.2009] [Citation(s) in RCA: 1087] [Impact Index Per Article: 77.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inwardly rectifying K(+) (Kir) channels allow K(+) to move more easily into rather than out of the cell. They have diverse physiological functions depending on their type and their location. There are seven Kir channel subfamilies that can be classified into four functional groups: classical Kir channels (Kir2.x) are constitutively active, G protein-gated Kir channels (Kir3.x) are regulated by G protein-coupled receptors, ATP-sensitive K(+) channels (Kir6.x) are tightly linked to cellular metabolism, and K(+) transport channels (Kir1.x, Kir4.x, Kir5.x, and Kir7.x). Inward rectification results from pore block by intracellular substances such as Mg(2+) and polyamines. Kir channel activity can be modulated by ions, phospholipids, and binding proteins. The basic building block of a Kir channel is made up of two transmembrane helices with cytoplasmic NH(2) and COOH termini and an extracellular loop which folds back to form the pore-lining ion selectivity filter. In vivo, functional Kir channels are composed of four such subunits which are either homo- or heterotetramers. Gene targeting and genetic analysis have linked Kir channel dysfunction to diverse pathologies. The crystal structure of different Kir channels is opening the way to understanding the structure-function relationships of this simple but diverse ion channel family.
Collapse
Affiliation(s)
- Hiroshi Hibino
- Department of Pharmacology, Graduate School of Medicine and The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
7
|
Song Z, Parker KJ, Enoh I, Zhao H, Olubajo O. Elucidation of spermidine interaction with nucleotide ATP by multiple NMR techniques. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2010; 48:123-128. [PMID: 19960498 PMCID: PMC2909874 DOI: 10.1002/mrc.2554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Interaction of polyamines with nucleotides plays a key role in many biological processes. Here we use multiple NMR techniques to characterize interaction of spermidine with adenosine 5'-triphosphate (ATP). Two-dimensional (1)H-(15)N spectra obtained from gs-HMBC experiments at varied pH show significant shift of N-1 peak around pH 2.0-7.0 range, suggesting that spermidine binds to N-1 site of ATP base. The binding facilitates N-1 deprotonation, shifting its pK(a) from 4.3 to 3.4. By correlating (15)N and (31)P chemical shift data, it is clear that spermidine is capable of concurrently binding to ATP base and phosphate sites around pH 4.0-7.0. The self-diffusion constants derived from (1)H PFG-diffusion measurements provide evidence that binding of spermidine to ATP is in 1:1 ratio, and pH variations do not induce significant nucleotide self-association in our samples. (31)P spectral analysis suggests that at neutral pH, Mg(2+) ion competes with spermidine and shows stronger binding to ATP phosphates. From (31)P kinetic measurements of myosin-catalyzed ATP hydrolysis, it is found that binding of spermidine affects the stability and reactivity of ATP. These NMR results are important for advancing the studies on nucleotide-polyamine interaction and its impact on nucleotide structures and activities under varied conditions.
Collapse
Affiliation(s)
- Zhiyan Song
- Department of Natural Sciences, Savannah State University, Savannah, GA 31404, USA.
| | | | | | | | | |
Collapse
|
8
|
Anumonwo JMB, Lopatin AN. Cardiac strong inward rectifier potassium channels. J Mol Cell Cardiol 2009; 48:45-54. [PMID: 19703462 DOI: 10.1016/j.yjmcc.2009.08.013] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 08/13/2009] [Accepted: 08/18/2009] [Indexed: 10/20/2022]
Abstract
Cardiac I(K1) and I(KACh) are the major potassium currents displaying classical strong inward rectification, a unique property that is critical for their roles in cardiac excitability. In the last 15 years, research on I(K1) and I(KACh) has been propelled by the cloning of the underlying inwardly rectifying potassium (Kir) channels, the discovery of the molecular mechanism of strong rectification and the linking of a number of disorders of cardiac excitability to defects in genes encoding Kir channels. Disease-causing mutations in Kir genes have been shown experimentally to affect one or more of the following channel properties: structure, assembly, trafficking, and regulation, with the ultimate effect of a gain- or a loss-of-function of the channel. It is now established that I(K1) and I(KACh) channels are heterotetramers of Kir2 and Kir3 subunits, respectively. Each homomeric Kir channel has distinct biophysical and regulatory properties, and individual Kir subunits often display different patterns of regional, cellular, and membrane distribution. These differences are thought to underlie important variations in the physiological properties of I(K1) and I(KACh). It has become increasingly clear that the contribution of I(K1) and I(KACh) channels to cardiac electrical activity goes beyond their long recognized role in the stabilization of resting membrane potential and shaping the late phase of action potential repolarization in individual myocytes but extends to being critical elements determining the overall electrical stability of the heart.
Collapse
Affiliation(s)
- Justus M B Anumonwo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109-5622, USA
| | | |
Collapse
|
9
|
Rubinstein M, Peleg S, Berlin S, Brass D, Dascal N. Galphai3 primes the G protein-activated K+ channels for activation by coexpressed Gbetagamma in intact Xenopus oocytes. J Physiol 2007; 581:17-32. [PMID: 17289785 PMCID: PMC2075207 DOI: 10.1113/jphysiol.2006.125864] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Accepted: 02/02/2007] [Indexed: 01/29/2023] Open
Abstract
G protein-activated K+ channels (GIRK) mediate postsynaptic inhibitory effects of neurotransmitters in the atrium and in the brain by coupling to G protein-coupled receptors (GPCRs). In neurotransmitter-dependent GIRK signalling, Gbetagamma is released from the heterotrimeric Galphabetagamma complex upon GPCR activation, activating the channel and attenuating its rectification. Now it becomes clear that Galpha is more than a mere Gbetagamma donor. We have proposed that Galphai3-GDP regulates GIRK gating, keeping its basal activity low but priming (predisposing) the channel for activation by agonist in intact cells, and by Gbetagamma in excised patches. Here we have further investigated GIRK priming by Galphai3 using a model in which the channel was activated by coexpression of Gbetagamma, and the currents were measured in intact Xenopus oocytes using the two-electrode voltage clamp technique. This method enables the bypass of GPCR activation during examination of the regulation of the channel in intact cells. Using this method, we further characterize the priming phenomenon. We tested and excluded the possibility that our estimates of priming are affected by artifacts caused by series resistance or large K+ fluxes. We demonstrate that both Galphai3 and membrane-attached Gbetagamma scavenger protein, m-phosducin, reduce the basal channel activity. However, Galphai3 allows robust channel activation by coexpressed Gbetagamma, in sharp contrast to m-phosducin, which causes a substantial reduction in the total Gbetagamma-induced current. Furthermore, Galphai3 also does not impair the Gbetagamma-dependent attenuation of the channel rectification, in contrast to m-phosducin, which prevents this Gbetagamma-induced modulation. The Galphai3-induced enhancement of direct activation of GIRK by Gbetagamma, demonstrated here for the first time in intact cells, strongly supports the hypothesis that Galphai regulates GIRK gating under physiological conditions.
Collapse
Affiliation(s)
- Moran Rubinstein
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Israel.
| | | | | | | | | |
Collapse
|
10
|
Abstract
The past 10 years of clinical experience have demonstrated the safety and efficacy of recombinant clotting factors. With the adoption of prophylactic strategies, there has been considerable progress in avoiding the complications of hemophilia. Now, insights from our understanding of clotting factor structure and function, mechanisms of hemophilia and inhibitors, gene therapy advances and a worldwide demand for clotting factor concentrates leave us on the brink of embracing targeted bioengineering strategies to further improve hemophilia therapeutics. The ability to bioengineer recombinant clotting factors with improved function holds promise to overcome some of the limitations in current treatment, the high costs of therapy and increase availability to a broader world hemophilia population. Most research has been directed at overcoming the inherent limitations of rFVIII expression and the inhibitor response. This includes techniques to improve rFVIII biosynthesis and secretion, functional activity, half-life and antigenicity/immunogenicity. Some of these proteins have already reached commercialization and have been utilized in gene therapy strategies, while others are being evaluated in pre-clinical studies. These novel proteins partnered with advances in gene transfer vector design and delivery may ultimately achieve persistent expression of FVIII leading to an effective long-term treatment strategy for hemophilia A. In addition, these novel FVIII proteins could be partnered with new advances in alternative recombinant protein production in transgenic animals yielding an affordable, more abundant supply of rFVIII. Novel rFIX proteins are being considered for gene therapy strategies whereas novel rVIIa proteins are being evaluated to improve the potency and extend their plasma half-life. This review will summarize the status of current recombinant clotting factors and the development and challenges of recombinant clotting factors bioengineered for improved function.
Collapse
Affiliation(s)
- S W Pipe
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
11
|
Affiliation(s)
- K Kurachi
- Age Dimension Research Center, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan.
| | | |
Collapse
|
12
|
Liu YL, Mingozzi F, Rodriguéz-Colôn SM, Joseph S, Dobrzynski E, Suzuki T, High KA, Herzog RW. Therapeutic Levels of Factor IX Expression Using a Muscle-Specific Promoter and Adeno-Associated Virus Serotype 1 Vector. Hum Gene Ther 2004; 15:783-92. [PMID: 15319035 DOI: 10.1089/1043034041648453] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Extensive studies in animal models of the X-linked bleeding disorder hemophilia B (deficiency in functional coagulation factor IX, F.IX) have shown that muscle-directed adeno-associated (AAV)-mediated F.IX gene transfer can be used to treat this disease. However, large vector doses of AAV-2 vector are required for therapeutic levels of expression, and the number of vector doses that can be injected per intramuscular site is limited. Several studies have shown that some of these limitations can be overcome by use of AAV serotype 1 vector. Here, we demonstrate levels of F.IX transgene expression from a synthetic muscle-specific promoter (C5-12) that were higher than from the cytomegalovirus (CMV) immediate-early enhancer-promoter in cultured muscle cells in vitro and approximately 50% of CMV-driven expression in vivo in murine skeletal muscle after AAV-1 gene transfer. These data show for the first time that a tissue-specific promoter can be used to achieve therapeutic levels of muscle-derived F.IX expression in the context of viral gene transfer. However, use of a muscle-specific promoter did not prevent antibody formation in response to a murine F.IX transgene product in mice with F.IX gene deletion, indicating that the risk of humoral immune responses remains in the context of an immunologically unfavorable mutation.
Collapse
Affiliation(s)
- Yi-Lin Liu
- Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Medical Center, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Kurachi Y, Ishii M. Cell signal control of the G protein-gated potassium channel and its subcellular localization. J Physiol 2004; 554:285-94. [PMID: 12923211 PMCID: PMC1664760 DOI: 10.1113/jphysiol.2003.048439] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
G protein-gated inward rectifier K(+) (K(G)) channels are directly activated by the betagamma subunits released from pertussis toxin-sensitive G proteins, and contribute to neurotransmitter-induced deceleration of heart beat, formation of slow inhibitory postsynaptic potentials in neurones and inhibition of hormone release in endocrine cells. The physiological roles of K(G) channels are critically determined by mechanisms which regulate their activity and their subcellular localization. K(G) channels are tetramers of inward rectifier K(+) (Kir) channel subunits, Kir3.x. The combination of Kir3.x subunits in each K(G) channel varies among tissues and cell types. Each subunit of the channel possesses one Gbetagamma binding site. The binding of Gbetagamma increases the number of functional K(G) channels via a mechanism that can be described by the Monod-Wyman-Changeux allosteric model. During voltage pulses K(G) channel current alters time dependently. The K(G) current exhibits inward rectification due to blockade of outward-going current by intracellular Mg(2+) and polyamines. Upon repolarization, this blockade is relieved practically instantaneously and then the current slowly increases further. This slow current alteration is called 'relaxation'. Relaxation is caused by the voltage-dependent behaviour of regulators of G protein signalling (RGS proteins), which accelerate intrinsic GTP hydrolysis mediated by the Galpha subunit. Thus, the relaxation behaviour of K(G) channels reflects the time course with which the G protein cycle is altered by RGS protein activity at each membrane potential. Subcellular localization of K(G) channels is controlled by several distinct mechanisms, some of which have been recently clarified. The neuronal K(G) channel, which contains Kir3.2c, is localized in the postsynaptic density (PSD) of various neurones including dopaminergic neurones in substantia nigra. Its localization at PSD may be controlled by PDZ domain-containing anchoring proteins. The K(G) channel in thyrotrophs is localized exclusively on secretary vesicles, which upon stimulation are rapidly inserted into the plasma membrane and causes hyperpolarization of the cell. This mechanism indicates a novel negative feedback regulation of exocytosis. In conclusion, K(G) channels are under the control of a variety of signalling molecules which regulate channel activity, subcellular localization and thus their physiological roles in myocytes, neurones and endocrine cells.
Collapse
Affiliation(s)
- Yoshihisa Kurachi
- Department of Pharmacology II, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | |
Collapse
|
14
|
De Marchi U, Pietrangeli P, Marcocci L, Mondovì B, Toninello A. l-Deprenyl as an inhibitor of menadione-induced permeability transition in liver mitochondria. Biochem Pharmacol 2003; 66:1749-54. [PMID: 14563485 DOI: 10.1016/s0006-2952(03)00474-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
L-Deprenyl, an inhibitor of mitochondrial monoamine oxidase B (MAO B), inhibits the swelling of liver mitochondria induced by the pro-oxidant 2-methyl-1,4-naphtoquinone with a K(i) dependent on quinone concentration. L-Deprenyl also inhibits the collapse of membrane potential, cation efflux, pyridine nucleotide oxidation and cytochrome c release, all events which accompany the osmotic change and are typical of membrane permeability transition induction, thus emphasizing the inhibitory effect of the drug on this phenomenon. Results show that this inhibition is not due to the effect of L-deprenyl on monoamine oxidase activity but is most likely due to a direct interaction of the drug with the pore forming structures. It is here proposed that L-deprenyl, being a propargylamine, at physiological pH has a protonated amino group able to interact with critical aromatic or anionic amino acidic residues. As a consequence, the opening of the transition pore is prevented. These results indicate a more generalized protective effect of L-deprenyl on mitochondrial functions, involving the inhibition of membrane permeability transition induced not only by the oxidation of substrates of MAO B, but also by pro-oxidant agents such as 2-methyl-1,4-naphtoquinone, which does not involve MAO B activity.
Collapse
Affiliation(s)
- Umberto De Marchi
- Dipartimento di Scienze Biomediche Sperimentali, Università di Padova, Padua, Italy
| | | | | | | | | |
Collapse
|
15
|
Abstract
Hemophilias A and B are X chromosome-linked bleeding disorders, which are mainly treated by repeated infusions of factor (F)VIII or FIX, respectively. In the present review, we specify the limitations in expression of recombinant (r)FVIII and summarize the bioengineering strategies that are currently being explored for constructing novel rFVIII molecules characterized by high efficiency expression and improved functional properties. We present the strategy to prolong FVIII lifetime by disrupting FVIII interaction with its clearance receptors and demonstrate how construction of human-porcine FVIII hybrid molecules can reduce their reactivity towards inhibitory antibodies. While the progress in improving rFIX is impeded by low recovery rates, the authors are optimistic that the efforts of basic science may ultimately lead to higher efficiency of replacement therapy of both hemophilias A and B.
Collapse
Affiliation(s)
- E L Saenko
- Department of Biochemistry, Jerome H. Holland Laboratory for the Biomedical Sciences, American Red Cross, Rockville, MD 20855, USA.
| | | | | | | | | |
Collapse
|
16
|
Plantier JL, Enjolras N, Rodriguez MHE, Massé JM, Cramer EM, Négrier C. The P-selectin cytoplasmic domain directs the cellular storage of a recombinant chimeric factor IX. J Thromb Haemost 2003; 1:292-9. [PMID: 12871503 DOI: 10.1046/j.1538-7836.2003.00071.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hemophilia B was recognized as a good candidate for gene therapy. Several strategies have been attempted and gave promising results in hemophilic animals but failed to achieve corrective levels in humans. To overcome this inconvenience we aimed to generate intracellular pools of factor (F)IX in cells that are implicated in the hemostatic response, e.g. endothelial cells and platelets. Upon stimulation, these cells release their granule content, which in this case would result in an increase in local FIX concentration, and could locally produce an effective hemostasis. In an attempt to produce an intracellular pool of releasable coagulation FIX, the cytoplasmic domain of the P-selectin (pselCT) molecule was fused to the carboxy-terminal extremity of the human FIX protein. The properties of this chimeric molecule (FIX-pselCT) were studied in AtT20, a cell line which possesses storage granules. As previously shown for transmembrane molecules but not for a soluble protein such as FIX, the pselCT fragment induces the storage of FIX-pselCT. The coagulant activity of FIX-pselCT was not affected by the addition of the pselCT tail. The treatment of AtT20 cells with different inhibitors revealed that FIX-pselCT was not submitted to intracellular degradation and that the half-life of the chimeric molecule was at least two times longer than that of FIX-WT. An immunoelectron microscopic analysis demonstrated a specific localization of FIX-pselCT within the ACTH-containing granules. Cell stimulation using Phorbol Myristrate Acetate (PMA), ionophore A-23187 or 8-Br-cAMP induced efficient release of an active FIX-pselCT. These data demonstrate that the addition of the cytoplasmic domain of P-selectin to FIX modifies the cellular fate of the FIX molecule by directing the recombinant protein toward regulated-secretory granules without altering its coagulant activity.
Collapse
Affiliation(s)
- J-L Plantier
- INSERM U331, Laboratoire d'Hémobiologie-Faculté de Médecine RTH, Laennec, Lyon, France
| | | | | | | | | | | |
Collapse
|
17
|
Hommers LG, Lohse MJ, Bünemann M. Regulation of the inward rectifying properties of G-protein-activated inwardly rectifying K+ (GIRK) channels by Gbeta gamma subunits. J Biol Chem 2003; 278:1037-43. [PMID: 12403784 DOI: 10.1074/jbc.m205325200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gbetagamma subunits are known to bind to and activate G-protein-activated inwardly rectifying K(+) channels (GIRK) by regulating their open probability and bursting behavior. Studying G-protein regulation of either native GIRK (I(KACh)) channels in feline atrial myocytes or heterologously expressed GIRK1/4 channels in Chinese hamster ovary cells and HEK 293 cells uncovered a novel Gbetagamma subunit mediated regulation of the inwardly rectifying properties of these channels. I(KACh) activated by submaximal concentrations of acetylcholine exhibited a approximately 2.5-fold stronger inward rectification than I(KACh) activated by saturating concentrations of acetylcholine. Similarly, the inward rectification of currents through GIRK1/4 channels expressed in HEK cells was substantially weakened upon maximal stimulation with co-expressed Gbetagamma subunits. Analysis of the outward current block underlying inward rectification demonstrated that the fraction of instantaneously blocked channels was reduced when Gbetagamma was over-expressed. The Gbetagamma induced weakening of inward rectification was associated with reduced potencies for Ba(2+) and Cs(+) to block channels from the extracellular side. Based on these results we propose that saturation of the channel with Gbetagamma leads to a conformational change within the pore of the channel that reduced the potency of extracellular cations to block the pore and increased the fraction of channels inert to a pore block in outward direction.
Collapse
Affiliation(s)
- Leif G Hommers
- University of Würzburg, Department of Pharmacology & Toxicology, Versbacherstrasse 9, Germany
| | | | | |
Collapse
|
18
|
Notley C, Killoran A, Cameron C, Wynd K, Hough C, Lillicrap D. The canine factor VIII 3'-untranslated region and a concatemeric hepatocyte nuclear factor 1 regulatory element enhance factor VIII transgene expression in vivo. Hum Gene Ther 2002; 13:1583-93. [PMID: 12228013 DOI: 10.1089/10430340260201671] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
If gene therapy is to be an effective treatment modality for hemophilia A, therapeutic levels and tissue-restricted expression of factor VIII (FVIII) must be achieved through optimization of transgene expression. To this end, we incorporated three types of sequence elements into a canine B domain-deleted FVIII transgene cassette and individually evaluated their effect on FVIII transgene expression. Functional FVIII activity was initially assessed in vitro and hydrodynamic injection of the different transgene constructs into mice was subsequently used as a model to compare in vivo expression of the various modified transgenes. Our results demonstrate that in vitro transgene expression is, in these studies, not a good predictor of in vivo transgene performance. In vivo analysis of a hybrid tissue-restricted promoter element, consisting of a concatemer of five hepatocyte nuclear factor 1 (HNF-1) consensus-binding motifs juxtaposed to the human FVIII proximal promoter, indicates that it is as efficient at mediating expression of the FVIII protein as the cytomegalovirus promoter. Addition of the full-length canine FVIII 3'-UTR also enhances transgene expression of FVIII in vivo. Sequence analysis of the canine FVIII 3'-UTR and human FVIII 3'-UTR indicates that the former lacks instability sequences and may therefore be more effective in stabilizing FVIII mRNA. Subsequent inclusion of FVIII introns 16 and 17 into the natural locations of the transgene disrupted mRNA processing and abolished expression of the FVIII protein. Introduction of intron 17 proximal to the FVIII cDNA did not enhance in vivo expression of canine FVIII from the transgene.
Collapse
Affiliation(s)
- Colleen Notley
- Department of Pathology, Richardson Laboratories, Queen's University, Kingston, Ontario, Canada K7L 3N6
| | | | | | | | | | | |
Collapse
|
19
|
Paulais M, Lourdel S, Teulon J. Properties of an inwardly rectifying K(+) channel in the basolateral membrane of mouse TAL. Am J Physiol Renal Physiol 2002; 282:F866-76. [PMID: 11934697 DOI: 10.1152/ajprenal.00238.2001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the properties of K(+) channels in the basolateral membrane of the cortical thick ascending limb (CTAL) using the patch-clamp technique. Approximately 34% of cell-attached patches contained an inwardly rectifying K(+) channel (K(+)-to-Na(+) permeability ratio approximately 22), having an inward conductance (G(in)) of 44 pS and an outward conductance (G(out)) of approximately 10 pS (G(in)/G(out) approximately 4). Channel activity (NP(o)) increased with depolarization. When the cytosolic sides of inside-out patches were exposed to an Mg(2+)-free medium, the channel had a G(in) of 50 pS and was weakly inwardly rectifying (G(in)/G(out) approximately 1). Cytosolic Mg(2+) reduced G(out), yielding a G(in)/G(out) of 3.8 at 1.3 mM Mg(2+). Internal Na(+) also yielded a G(in)/G(out) of 1.6 at 20 mM Na(+). Spermine reduced NP(o) on inside-out membrane patches. Sensitivity to spermine at depolarizing voltages [half-maximal inhibitory concentration (K(i)) = 0.2 microM] was much greater than at hyperpolarizing voltages (K(i) = 26 microM). Half-inactivation by 0.5 microM spermine occurred at a clamp potential of 43 mV, with an effective valence of 1.25. A sigmoid relationship between bath pH and NP(o) of inside-out membrane patches was observed, with a pK of 7.6 and a Hill coefficient of 1.8. Intracellular acidification also reduced the NP(o) of cell-attached patches. This channel is probably a major component of K(+) conductance in the CTAL basolateral membrane.
Collapse
Affiliation(s)
- Marc Paulais
- Institut National de la Santé et de la Recherche Médicale U.426, Institut Fédératif de Recherche 02, Faculté de Médecine Xavier Bichat, Université Paris 7, 75018 Paris, France.
| | | | | |
Collapse
|
20
|
Lourdel S, Paulais M, Cluzeaud F, Bens M, Tanemoto M, Kurachi Y, Vandewalle A, Teulon J. An inward rectifier K(+) channel at the basolateral membrane of the mouse distal convoluted tubule: similarities with Kir4-Kir5.1 heteromeric channels. J Physiol 2002; 538:391-404. [PMID: 11790808 PMCID: PMC2290070 DOI: 10.1113/jphysiol.2001.012961] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In this study, K(+) channels present in the basolateral membrane of the distal convoluted tubule (DCT) were investigated using patch-clamp methods. In addition, Kir4.1, Kir4.2 and Kir5.1 inward rectifier channels were investigated using RT-PCR and immunohistochemistry (Kir4.1). DCTs were microdissected from collagenase-treated mouse kidneys. One type of K(+) channel was detected in about 50 % of cell-attached patches from the DCT basolateral membrane; this channel was inwardly rectifying and had an inward conductance (g(in)) of approximately 40 pS at an external [K(+)] of 145 mM. The current-voltage relationship was linear when inside-out patches were exposed to a Mg(2+)-free medium. Mg(2+) at a concentration of 1.2 mM considerably reduced the outward conductance (g(out)), yielding a g(in)/g(out) ratio of approximately 4.7. The polycation spermine (5 x 10(-7) M) reduced the open probability (P(o)) by 50 %. Channel activity was dependent upon the intracellular pH, with acid pH decreasing, and basic pH increasing, P(o). Internal ATP (2 mM) and Ca(2+) (up to 10(-3) M) had no effect. Channel activity declined irreversibly when the inner side of the patch was exposed to Mg(2+). Kir4.1, Kir4.2 and Kir5.1 mRNAs were all detected in the DCT. The Kir4.1 protein co-localised with the Na(+)-Cl(-) cotransporter, which is specific to the DCT, and was located on basolateral membranes. The DCT K(+) channel differs from other functionally identified renal K(+) channels with regard to its inhibition by spermine and insensitivity to internal ATP and Ca(2+). At the current state of knowledge, the channel is similar to Kir4.1-Kir5.1 and Kir4.2-Kir5.1 heteromeric channels, but not to Kir4.1 or Kir4.2 homomeric channels.
Collapse
Affiliation(s)
- Stéphane Lourdel
- INSERM U426, Institut Fédératif de Recherche 02, Faculté de Médecine Xavier Bichat, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Bender K, Wellner-Kienitz MC, Inanobe A, Meyer T, Kurachi Y, Pott L. Overexpression of monomeric and multimeric GIRK4 subunits in rat atrial myocytes removes fast desensitization and reduces inward rectification of muscarinic K(+) current (I(K(ACh))). Evidence for functional homomeric GIRK4 channels. J Biol Chem 2001; 276:28873-80. [PMID: 11384974 DOI: 10.1074/jbc.m102328200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
K(+) channels composed of G-protein-coupled inwardly rectifying K(+) channel (GIRK) (Kir3.0) subunits are expressed in cardiac, neuronal, and various endocrine tissues. They are involved in inhibiting excitability and contribute to regulating important physiological functions such as cardiac frequency and secretion of hormones. The functional cardiac (K((ACh))) channel activated by G(i)/G(o)-coupled receptors such as muscarinic M(2) or purinergic A(1) receptors is supposed to be composed of the subunits GIRK1 and GIRK4 in a heterotetrameric (2:2) fashion. In the present study, we have manipulated the subunit composition of the K((ACh)) channels in cultured atrial myocytes from hearts of adult rats by transient transfection of vectors encoding for GIRK1 or GIRK4 subunits or GIRK4 concatemeric constructs and investigated the effects on properties of macroscopic I(K(ACh)). Transfection with a GIRK1 vector did not cause any measurable effect on properties of I(K(ACh)), whereas transfection with a GIRK4 vector resulted in a complete loss in desensitization, a reduction of inward rectification, and a slowing of activation. Transfection of myocytes with a construct encoding for a concatemeric GIRK4(2) subunit had similar effects on desensitization and inward rectification. Following transfection of a tetrameric construct (GIRK4(4)), these changes in properties of I(K(ACh)) were still observed but were less pronounced. Heterologous expression in Chinese hamster ovary cells and human embryonic kidney 293 cells of monomeric, dimeric, and tetrameric GIRK4 resulted in robust currents activated by co-expressed A(1) and M(2) receptors, respectively. These data provide strong evidence that homomeric GIRK4 complexes form functional G(beta)gamma gated ion channels and that kinetic properties of GIRK channels, such as activation rate, desensitization, and inward rectification, depend on subunit composition.
Collapse
Affiliation(s)
- K Bender
- Institut für Physiologie, Ruhr-Universität Bochum, D-4480 Bochum, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
The cardiac inward rectifier potassium current (I(K1)), present in all ventricular and atrial myocytes, has been suggested to play a major role in repolarization of the action potential and stabilization of the resting potential. The molecular basis is now ascribed to members of the Kir2 sub-family of inward rectifier K channel genes, and the availability of recombinant expression systems has led to elucidation of the mechanism of inward rectification, as well as additional regulatory mechanisms involving intracellular pH and phosphorylation. In vivo manipulation of the genes encoding I(K1)and regulatory proteins now promise to provide new insights to the role of this conductance in the heart. This review details recent advances and considers the prospects for further elucidation of the role of this conductance in cardiac electrical activity.
Collapse
Affiliation(s)
- A N Lopatin
- Department of Physiology, University of Michigan, 1150 W Medical Center Drive, Ann Arbor, MI 48109-0622, USA
| | | |
Collapse
|
23
|
Metzler DE, Metzler CM, Sauke DJ. Transferring Groups by Displacement Reactions. Biochemistry 2001. [DOI: 10.1016/b978-012492543-4/50015-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
24
|
Lancaster MK, Dibb KM, Quinn CC, Leach R, Lee JK, Findlay JB, Boyett MR. Residues and mechanisms for slow activation and Ba2+ block of the cardiac muscarinic K+ channel, Kir3.1/Kir3.4. J Biol Chem 2000; 275:35831-9. [PMID: 10956662 DOI: 10.1074/jbc.m006565200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mechanisms and residues responsible for slow activation and Ba(2+) block of the cardiac muscarinic K(+) channel, Kir3.1/Kir3.4, were investigated using site-directed mutagenesis. Mutagenesis of negatively charged residues located throughout the pore of the channel (in H5, M2, and proximal C terminus) reduced or abolished slow activation. The strongest effects resulted from mutagenesis of residues in H5 close to the selectivity filter; mutagenesis of residues in M2 and proximal C terminus equivalent to those identified as important determinants of the activation kinetics of Kir2.1 was less effective. In giant patches, slow activation was present in cell-attached patches, lost on excision of the patch, and restored on perfusion with polyamine. Mutagenesis of residues in H5 and M2 close to the selectivity filter also decreased Ba(2+) block of the channel. A critical residue for Ba(2+) block was identified in Kir3.4. Mutagenesis of the equivalent residue in Kir3.1 failed to have as pronounced an effect on Ba(2+) block, suggesting an asymmetry of the channel pore. It is concluded that slow activation is principally the result of unbinding of polyamines from negatively charged residues close to the selectivity filter of the channel and not an intrinsic gating mechanism. Ba(2+) block involves an interaction with the same residues.
Collapse
Affiliation(s)
- M K Lancaster
- School of Biomedical Sciences and Biochemistry & Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
25
|
Lopatin AN, Shantz LM, Mackintosh CA, Nichols CG, Pegg AE. Modulation of potassium channels in the hearts of transgenic and mutant mice with altered polyamine biosynthesis. J Mol Cell Cardiol 2000; 32:2007-24. [PMID: 11040105 DOI: 10.1006/jmcc.2000.1232] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inward rectification of cardiac I(K1)channels was modulated by genetic manipulation of the naturally occurring polyamines. Ornithine decarboxylase (ODC) was overexpressed in mouse heart under control of the cardiac alpha -myosin heavy chain promoter (alpha MHC). In ODC transgenic hearts, putrescine and cadaverine levels were highly elevated ( identical with 35-fold for putrescine), spermidine was increased 3.6-fold, but spermine was essentially unchanged. I(K1)density was reduced by identical with 38%, although the voltage-dependence of rectification was essentially unchanged. Interestingly, the fast component of transient outward (I(to,f)) current was increased, but the total outward current amplitude was unchanged. I(K1)and I(to)currents were also studied in myocytes from mutant Gyro (Gy) mice in which the spermine synthase gene is disrupted, leading to a complete loss of spermine. I(K1)current densities were not altered in Gy myocytes, but the steepness of rectification was reduced indicating a role for spermine in controlling rectification. Intracellular dialysis of myocytes with putrescine, spermidine and spermine caused reduction, no change and increase of the steepness of rectification, respectively. Taken together with kinetic analysis of I(K1)activation these results are consistent with spermine being a major rectifying factor at potentials positive to E(K), spermidine dominating at potentials around and negative to E(K), and putrescine playing no significant role in rectification in the mouse heart.
Collapse
Affiliation(s)
- A N Lopatin
- Department of Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
26
|
Fujita S, Inanobe A, Chachin M, Aizawa Y, Kurachi Y. A regulator of G protein signalling (RGS) protein confers agonist-dependent relaxation gating to a G protein-gated K+ channel. J Physiol 2000; 526 Pt 2:341-7. [PMID: 10896722 PMCID: PMC2270023 DOI: 10.1111/j.1469-7793.2000.00341.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
1. The effects of RGS4 on the voltage-dependent relaxation of G protein-gated K+ (KG) channels were examined by heterologous expression in Xenopus oocytes. 2. While the relaxation kinetics was unaffected by the acetylcholine concentration ([ACh]) in the absence of RGS4, it became dependent on [ACh] when RGS4 was co-expressed. 3. Kinetic analyses indicated that RGS4 confers to the KG channel a voltage-independent inhibitory gating mechanism, which was attenuated by ACh in a concentration-dependent fashion. 4. In vitro biochemical studies showed that RGS4 could bind to the protein complex containing KG channel subunits. 5. Since the native cardiac KG channel exhibited similar agonist-dependent relaxation kinetics to that mediated by RGS4, it is suggested that KG channel gating is a novel physiological target of RGS protein-mediated regulation.
Collapse
Affiliation(s)
- S Fujita
- Department of Pharmacology II, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | |
Collapse
|
27
|
Properties, regulation, and role of potassium channels of smooth muscle. ACTA ACUST UNITED AC 2000. [DOI: 10.1016/s1569-2590(00)08010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
28
|
De Geest B, Van Linthout S, Lox M, Collen D, Holvoet P. Sustained expression of human apolipoprotein A-I after adenoviral gene transfer in C57BL/6 mice: role of apolipoprotein A-I promoter, apolipoprotein A-I introns, and human apolipoprotein E enhancer. Hum Gene Ther 2000; 11:101-12. [PMID: 10646643 DOI: 10.1089/10430340050016193] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Elevation of HDL cholesterol, after adenoviral apolipoprotein A-I (apo A-I) gene transfer, may delay or revert ischemic cardiovascular disease, provided transgene expression is persistent. Previously, we observed transient human apo A-I expression after adenoviral gene transfer with a cytomegalovirus (CMV)-driven construct containing the human apo A-I cDNA. Therefore, the effects of promoters (CMV or 256 base pairs of the human apo A-I promoter), introns of the human apo A-I gene, and the liver-specific human apolipoprotein E (apo E) enhancer on adenovirus-mediated human apo A-I expression were evaluated in C57BL/6 mice. In the presence of the CMV promoter, human apo A-I introns prolonged expression above 20 mg/dl from 14 to 35 days. Addition of one, two, or four copies of the human apo E enhancer in these constructs resulted in a copy-dependent but transient increase in expression for 14 days. The apo A-I promoter induced 3.2-fold lower peak levels of human apo A-I than did the CMV promoter, but insertion of four apo E enhancers in the apo A-I promoter-driven construct resulted in human apo A-I levels above 20 mg/dl for 6 months. The decline between day 6 and day 35 of human apo A-I expression driven by the CMV promoter was due to (1) a 2.5-fold decline in transgene DNA levels that is not observed with apo A-I promoter-driven constructs, and (2) CMV promoter attenuation as evidenced by a 7.6-fold decline in the human apo A-I mRNA/human apo A-I DNA copy number ratio between day 6 and day 35. Hepatotoxicity, as evidenced by up to 10-fold higher serum levels of transaminases on day 6 after gene transfer with CMV promoter-driven constructs than with apo A-I promoter-driven constructs, probably caused the accelerated decline of transgene DNA. In conclusion, gene transfer with an adenovirus comprising the 256-bp apo A-I promoter, the genomic apo A-I DNA, and four apo E enhancers, all of human origin, is associated with low hepatotoxicity and with the absence of promoter shutoff resulting in human apo A-I expression above 20 mg/dl for up to 6 months.
Collapse
Affiliation(s)
- B De Geest
- Center for Molecular and Vascular Biology, Leuven, Belgium
| | | | | | | | | |
Collapse
|
29
|
Abstract
The aim of this review is to provide basic information on the electrophysiological changes during acute ischemia and reperfusion from the level of ion channels up to the level of multicellular preparations. After an introduction, section II provides a general description of the ion channels and electrogenic transporters present in the heart, more specifically in the plasma membrane, in intracellular organelles of the sarcoplasmic reticulum and mitochondria, and in the gap junctions. The description is restricted to activation and permeation characterisitics, while modulation is incorporated in section III. This section (ischemic syndromes) describes the biochemical (lipids, radicals, hormones, neurotransmitters, metabolites) and ion concentration changes, the mechanisms involved, and the effect on channels and cells. Section IV (electrical changes and arrhythmias) is subdivided in two parts, with first a description of the electrical changes at the cellular and multicellular level, followed by an analysis of arrhythmias during ischemia and reperfusion. The last short section suggests possible developments in the study of ischemia-related phenomena.
Collapse
Affiliation(s)
- E Carmeliet
- Centre for Experimental Surgery and Anesthesiology, University of Leuven, Leuven, Belgium
| |
Collapse
|
30
|
Kawamura S, Kurachi S, Deyashiki Y, Kurachi K. Complete nucleotide sequence, origin of isoform and functional characterization of the mouse hepsin gene. EUROPEAN JOURNAL OF BIOCHEMISTRY 1999; 262:755-64. [PMID: 10411637 DOI: 10.1046/j.1432-1327.1999.00431.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hepsin, a type-II membrane-associated serine protease, has been implicated in cell growth and development as well as possible initiation of blood coagulation. Here, we report on the complete nucleotide sequence, functional characterization of key structural features and the promoter of the mouse hepsin gene. The gene has a size of approximately 17 kb, and is composed of 12, 13, or 14 exons depending on alternative intron splicings - one in the 5'-UTR and the other two in the second intron. The latter two, which occur in approximately half of the hepsin transcripts, generate a hepsin mRNA species with an extra exon, which is responsible for producing a hepsin isoform with a unique 20-residue sequence inserted in the cytoplasmic portion of hepsin. Most hepsin transcripts have the 5'-UTR intron spliced, and its splicing can occur independently of the other alternative splicings. The transcriptional initiation site was determined to be 636 bp upstream of the first ATG site in a cytidine-rich region. The 5'-flanking region of hepsin up to nucleotide 274 showed a substantial promoter activity in HepG2 cells, with its expression activity sevenfold higher in the presence of the 5'-UTR intron sequence in comparison to that without the intron sequence. The basal promoter region contains potential binding sites for several transcription factors including SP1, AP2, C/EBP, LF-A1, and E box, which may be responsible for ubiquitous, but liver- and kidney-preferred tissue expression of the hepsin gene.
Collapse
Affiliation(s)
- S Kawamura
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor 48109-0618, USA
| | | | | | | |
Collapse
|
31
|
Laschet J, Trottier S, Leviel V, Guibert B, Bansard JY, Chauvel P, Bureau M. Heterogeneous distribution of polyamines in temporal lobe epilepsy. Epilepsy Res 1999; 35:161-72. [PMID: 10372569 DOI: 10.1016/s0920-1211(99)00009-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Polyamine contents were determined in human temporal lobe epilepsy. In the seven patients studied, stereoelectroencephalography (SEEG) located the epileptogenic focus in Ammon's horn and neuropathological findings were limited to hippocampal gliosis and sclerosis. Each polyamine exhibited a specific regional distribution. The most important variations were observed for spermidine and spermine while putrescine levels varied less. The regional variation was predominant in middle > posterior > anterior parts of the temporal lobe. Spermine contents and the spermidine/spermine (SPD/SPM) index varied especially in the middle and posterior parts of the hippocampus. Metabolic SPD/SPM index and spermidine levels were found to be drastically increased in almost all limbic parts when compared to neocortical regions. The opposite was observed for spermine. The heterogeneous distribution of polyamines was compared to abnormal electrical activities recorded by SEEG: SPD/SPM index and spermidine levels were sharply increased in seizure onset areas and high levels of spermine were detected in temporal cortex propagation areas. The presently reported heterogeneity of polyamine contents might contribute to modulate differentially the local control of excitability in human temporal epilepsy.
Collapse
Affiliation(s)
- J Laschet
- Laboratory of Neurosciences, School of Medicine, University of Rennes, France.
| | | | | | | | | | | | | |
Collapse
|
32
|
Wellner-Kienitz MC, Bender K, Brandts B, Meyer T, Pott L. Antisense oligonucleotides against receptor kinase GRK2 disrupt target selectivity of beta-adrenergic receptors in atrial myocytes. FEBS Lett 1999; 451:279-83. [PMID: 10371205 DOI: 10.1016/s0014-5793(99)00594-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
K+ channels composed of GIRK subunits are predominantly expressed in the heart and various regions of the brain. They are activated by betagamma-subunits released from pertussis toxin-sensitive G-proteins coupled to different seven-helix receptors. In rat atrial myocytes, activation of K(ACh) channels is strictly limited to receptors coupled to pertussis toxin-sensitive G-proteins. Upon treatment of myocytes with antisense oligodesoxynucleotides against GRK2, a receptor kinase with Gbetagamma binding sites, in a fraction of cells, K(ACh) channels can be activated by beta-adrenergic receptors. Sensitivity to beta-agonist is insensitive to pertussis toxin treatment. These findings demonstrate a potential role of Gbetagamma binding proteins for target selectivity of G-protein-coupled receptors.
Collapse
Affiliation(s)
- M C Wellner-Kienitz
- Institut für Physiologie, Abteilung Zelluläre Physiologie, Ruhr-Universität Bochum, Germany
| | | | | | | | | |
Collapse
|
33
|
Hortelano G, Xu N, Vandenberg A, Solera J, Chang PL, Ofosu FA. Persistent delivery of factor IX in mice: gene therapy for hemophilia using implantable microcapsules. Hum Gene Ther 1999; 10:1281-8. [PMID: 10365659 DOI: 10.1089/10430349950017969] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Severe hemophilia B is a life-threatening, life long condition caused by absence of or defective coagulation factor IX. Gene therapy could provide an alternative treatment to repeated injection of plasma-derived concentrate or recombinant factor IX. We have previously described the use of implantable microcapsules containing recombinant myoblasts to deliver human factor IX in mice. This study reports the generation of improved myoblast-specific expression vectors. Mouse myoblast clones transfected with the various vectors secreted factor IX in vitro, at rates between 70 and 1000 ng/10(6) cells/day. The recombinant myoblast clones were then encapsulated and implanted into mice. Immunocompetent mice implanted with encapsulated myoblasts had up to 65 ng of factor IX per milliliter in their plasma for up to 14 days, after which antibodies to human factor IX became detectable, and this coincided with decreased factor IX in mouse plasma. In immunodeficient mice, however, factor IX delivery was maintained at a constant level for at least 6 weeks (end of experiment). Interestingly, the highest-secreting myoblast clone in vitro did not deliver the highest level of hFIX in vivo. This discrepancy observed between performance in vitro and in vivo may have important implications for the development of gene therapy protocols based on recombinant cells.
Collapse
Affiliation(s)
- G Hortelano
- Department of Pathology and Molecular Medicine, Canadian Blood Services, McMaster University, Hamilton, Ontario
| | | | | | | | | | | |
Collapse
|
34
|
Wahlfors JJ, Morgan RA. Production of minigene-containing retroviral vectors using an alphavirus/retrovirus hybrid vector system. Hum Gene Ther 1999; 10:1197-206. [PMID: 10340551 DOI: 10.1089/10430349950018184] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In an attempt to increase the synthesis of human clotting factors VIII and IX in transduced cells, optimized expression cassettes containing genomic genelike elements (minigenes) were assembled. Plasmid DNA containing factor VIII or factor IX minigenes and driven by three human cellular promoters (albumin, factor IX, PGK) or the strong viral promoter RSV-LTR were electroporated into TE671 and HepG2 cell lines, and clotting factor levels were determined by ELISA. In comparison with a parallel transfection of MLV-LTR-promoted retroviral vector plasmid DNAs, the PGK- and RSV-LTR-promoted minigene constructs produced equal or greater amounts of clotting factor proteins. A factor IX minigene cassette was cloned into the retrovirus-based gene transfer vector LN (in both forward and reverse orientations) and the minigene vector was introduced into the Phoenix retroviral packaging cell line. Analysis of neo(r) cells demonstrated that insertion of a factor IX minigene into the retroviral vector LN resulted in rearrangement of the factor IX sequence and loss of factor IX expression in the Phoenix packaging cell line. The same factor IX minigene was then inserted into an alphavirus/retrovirus hybrid vector that facilitates the synthesis of retroviral vector RNA in the cytoplasm of cells. Alphavirus/retrovirus virions were produced and used to transduce the Phoenix retroviral vector packaging cell line. The cytoplasmically produced factor IX minigene-containing retroviral vectors were collected and used to transduce TE671 cells. Analysis of transduced cells demonstrated stable transfer of the minigene and expression of factor IX.
Collapse
Affiliation(s)
- J J Wahlfors
- Clinical Gene Therapy Branch/National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-1851, USA
| | | |
Collapse
|
35
|
Chapter 10 Inwardly Rectifying Potassium Channels: Mechanisms of Rectification. CURRENT TOPICS IN MEMBRANES 1999. [DOI: 10.1016/s0070-2161(08)60926-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
36
|
Chapter 14 Regulation of Ion Channels by Membrane Proteins. CURRENT TOPICS IN MEMBRANES 1999. [DOI: 10.1016/s0070-2161(08)60930-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
37
|
Sorota S, Chlenov M, Du XY, Kagan M. ATP-dependent activation of the atrial acetylcholine-induced K+ channel does not require nucleoside diphosphate kinase activity. Circ Res 1998; 82:971-9. [PMID: 9598594 DOI: 10.1161/01.res.82.9.971] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prior reports by others have shown that cytoplasmically applied ATP can activate the acetylcholine-induced K+ channel in inside-out atrial membrane patches when no guanine nucleotides are present in the solution bathing the cytosolic face of the membrane. A nucleoside diphosphate kinase mechanism was proposed to explain the activation by ATP. We show in the present study that cytoplasmic adenylylimidodiphosphate mimics the activation by ATP. Unlike ATP, the activation by adenylylimidodiphosphate does not subside on washout. Although commercially available adenylylimidodiphosphate is contaminated by guanylylimidodiphosphate, the activation by adenylylimidodiphosphate still occurs after HPLC purification to remove guanine nucleotide contamination. Adenylylimidodiphosphate does not support phosphotransferase activity by nucleoside diphosphate kinase. Therefore, nucleoside diphosphate kinase activity cannot explain the activation of atrial acetylcholine-induced K+ current by ATP and adenylylimidodiphosphate. We hypothesize that the activation by millimolar concentrations of ATP is due to binding of adenine nucleotide to the guanine nucleotide binding site of the G protein(s) responsible for stimulating the acetylcholine-induced K+ current.
Collapse
Affiliation(s)
- S Sorota
- Department of Pharmacology, Columbia University, New York, NY 10032, USA.
| | | | | | | |
Collapse
|
38
|
Changes in expression of the ion channels in mammalian cardiomyocytes in early embryogenesis. NEUROPHYSIOLOGY+ 1998. [DOI: 10.1007/bf02463432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Niu XW, Meech RW. The effect of polyamines on KATP channels in guinea-pig ventricular myocytes. J Physiol 1998; 508 ( Pt 2):401-11. [PMID: 9508805 PMCID: PMC2230875 DOI: 10.1111/j.1469-7793.1998.401bq.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
1. The effect of natural polyamines on KATP channels was studied using inside-out patches from guinea-pig ventricular myocytes. 2. At a holding potential of +40 mV, spermine at the intracellular membrane surface reduced the KATP channel open probability (Popen) in a dose-dependent manner. Half-maximal inhibition occurred at 29 microM with a Hill coefficient of 1.2. 3. The effect of spermine on Popen was not greatly influenced by the membrane potential but there appeared to be a small reduction in unitary current amplitude during strong depolarizations. 4. Analysis of KATP single channel kinetics showed that spermine inhibited the channel by decreasing the mean open time and introducing transitions to a long closed state. 5. Spermidine (0.1 mM) was found to have a similar effect to spermine. Putrescine (10 mM) was found to block more effectively at positive membrane potentials. Up to 20 mM arginine had no significant effect on KATP channels. 6. Our results indicate that natural polyamines influence native KATP channel gating in cardiac myocytes.
Collapse
Affiliation(s)
- X W Niu
- Department of Physiology, School of Veterinary Science, University of Bristol, Bristol BS2 8EJ, UK
| | | |
Collapse
|
40
|
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
|
41
|
Wahlfors JJ, Xanthopoulos KG, Morgan RA. Semliki Forest virus-mediated production of retroviral vector RNA in retroviral packaging cells. Hum Gene Ther 1997; 8:2031-41. [PMID: 9414252 DOI: 10.1089/hum.1997.8.17-2031] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Retroviral vectors are efficient tools for gene transfer studies. Their major advantage is that they can permanently integrate the transgene into the target cell's genome. However, because of the compulsory nuclear expression phase of their life cycle, it can be difficult for retroviruses to carry complex expression cassettes. In a attempt to mimic the structural features of most eukaryotic genes and obtain a potentially self-amplifying system for retrovirus production, we tested the feasibility of Semliki Forest virus (SFV) expression to mediate cytoplasmic synthesis of retrovirus vector RNA. An equivalent of a retrovirus virion RNA (retrovirus vector cassette, RVC) was cloned under the SFV 26S promoter, and full-length chimeric SFV-RVC RNA was produced in vitro. This RNA was introduced into retrovirus packaging cells, either via electroporation or transduction in SFV virions, and supernatants were analyzed for the presence of biologically active retroviruses. We demonstrate that this strategy can be used for cytoplasmic retrovirus production. The resulting viral particles are fully functional; they can transduce target cells, undergo reverse transcription, and integrate into genomic DNA. We also demonstrate that the SFV virion-based RVC delivery into packaging cells can yield high transient titers, in this case more than 10(5) G418R cfu/ml. This study shows that a simple, one-plasmid, heterologous viral RNA production system can be used to create functional retroviral RNA outside the cell nucleus.
Collapse
Affiliation(s)
- J J Wahlfors
- Clinical Gene Therapy Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
42
|
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
|
43
|
Abstract
Endogenous polyamines, in particular spermine, have been found to cause block and modulation of a number of types of ion channel. Intracellular spermine is responsible for intrinsic gating and rectification of strong inward rectifier K+ channels by directly plugging the ion channel pore. These K+ channels control the resting membrane potential in both excitable and non-excitable cells, and control the excitability threshold in neurons and muscle cells. Intracellular spermine causes inward rectification at some subtypes of Ca2+-permeable glutamate receptors in the central nervous system, again by plugging the receptor channel pore, and spermine can even permeate the ion channel of these receptors. Extracellular spermine has multiple effects at the N-methyl-d-aspartate (NMDA) subtype of glutamate receptor, including stimulation that increases the size of NMDA receptor currents, and voltage-dependent block. A number of polyamine-conjugated arthropod toxins and synthetic polyamine analogues are potent antagonists of glutamate receptors, and represent new tools with which to study these receptors. Interactions of polyamines with other types of cation channels have been reported. This area of research represents a new biology and a new pharmacology of polyamines.
Collapse
Affiliation(s)
- K Williams
- Department of Pharmacology, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104-6084, USA
| |
Collapse
|
44
|
Abstract
The endogenous polyamines, spermine, spermidine, and putrescine have effects on several types of cation channels. Intracellular polyamines, in particular spermine, contribute to intrinsic gating and rectification of strong inward rectifier K+ channels. Intracellular spermine is also responsible for inward rectification of some types of Ca(2+)-permeable AMPA and kainate receptors. Spermine has a number of effects on the activity of the NMDA subtype of glutamate receptor, involving two or more extracellular polyamine binding sites on the NMDA receptor. In K+ channels and glutamate receptors, some of the amino acids in the receptor/channel structure that influence to polyamines have been identified, leading to a partial understanding of the effects of polyamines at a molecular level. Block of K+ channels by intracellular polyamines is likely to be an important receptors by intracellular spermine and modulation by extracellular spermine may affect excitability and the influx of Ca2+ in neurons and glial cells of the nervous system.
Collapse
Affiliation(s)
- K Williams
- Department of Pharmacology, University of Pennsylvania, School of Medicine, Philadelphia 19104-6084, USA
| |
Collapse
|
45
|
Abstract
Potassium channels contribute to the excitability of neurons and signaling in the nervous system. They arise from multiple gene families including one for voltage-gated potassium channels and one for inwardly rectifying potassium channels. Features of potassium permeation, channel gating and regulation, and subunit interaction have been analyzed. Potassium channels of similar design have been found in animals ranging from jellyfish to humans, as well as in plants, yeast, and bacteria. Structural similarities are evident for the pore-forming alpha subunits and for the beta subunits, which could potentially regulate channel activity according to the level of energy and/or reducing power of the cell.
Collapse
Affiliation(s)
- L Y Jan
- Department of Physiology, University of California, San Francisco 94143-0724, USA
| | | |
Collapse
|
46
|
Abstract
GABA and the GABAB receptor agonist baclofen activated a potassium conductance in acutely dissociated hippocampal CA3 neurons. Baclofen-activated current required internal GTP, was purely potassium selective, and showed strong inward rectification. As with acetylcholine-activated current in atrial myocytes, external Cs+ blocked inward but not outward current in a highly voltage-dependent manner, whereas Ba2+ blocked with no voltage dependence. Unlike the cardiac current, however, the baclofen-activated current showed no intrinsic voltage-dependent relaxation. With fast solution exchange, current was activated by baclofen or GABA with a lag of approximately 50 msec followed by an exponential phase (time constant approximately 225 msec at saturating agonist concentrations); deactivation was preceded by a lag of approximately 150 msec and occurred with a time constant of approximately 1 sec. GABA activated the potassium conductance with a half maximally effective concentration (EC50) of 1.6 microM, much lower than that for activation of GABAA receptor-activated chloride current in the same cells (EC50 approximately 25 microM). At low GABA concentrations, activation of the GABAB current had a Hill coefficient of 1.4-2.1, suggesting cooperativity in the receptor-to-channel pathway. Although the maximal conductance activated by GABAB receptors is much smaller than that activated by GABAA receptors, its higher sensitivity to GABA and slower time course make it well suited to respond to low concentrations of extra-synaptic GABA.
Collapse
|
47
|
Shyng SL, Sha Q, Ferrigni T, Lopatin AN, Nichols CG. Depletion of intracellular polyamines relieves inward rectification of potassium channels. Proc Natl Acad Sci U S A 1996; 93:12014-9. [PMID: 8876254 PMCID: PMC38175 DOI: 10.1073/pnas.93.21.12014] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Two different approaches were used to examine the in vivo role of polyamines in causing inward rectification of potassium channels. In two-microelectrode voltage-clamp experiments, 24-hr incubation of Xenopus oocytes injected with 50 nl of difluoromethylornithine (5 mM) and methylglyoxal bis(guanylhydrazone) (1 mM) caused an approximate doubling of expressed Kir2.1 currents and relieved rectification by causing an approximately +10-mV shift of the voltage at which currents are half-maximally inhibited. Second, a putrescine auxotrophic, ornithine decarboxylase-deficient Chinese hamster ovary (O-CHO) cell line was stably transfected with the cDNA encoding Kir2.3. Withdrawal of putrescine from the medium led to rapid (1-day) loss of the instantaneous phase of Kir2.3 channel activation, consistent with a decline of intracellular putrescine levels. Four days after putrescine withdrawal, macroscopic conductance, assessed using an 86Rb+ flux assay, was approximately doubled, and this corresponded to a +30-mV shift of V1/2 of rectification. With increasing time after putrescine withdrawal, there was an increase in the slowest phase of current activation, corresponding to an increase in the spermine-to-spermidine ratio over time. These results provide direct evidence for a role of each polyamine in induction of rectification, and they further demonstrate that in vivo modulation of rectification is possible by manipulation of polyamine levels using genetic and pharmacological approaches.
Collapse
Affiliation(s)
- S L Shyng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
48
|
Wang JM, Zheng H, Sugahara Y, Tan J, Yao SN, Olson E, Kurachi K. Construction of human factor IX expression vectors in retroviral vector frames optimized for muscle cells. Hum Gene Ther 1996; 7:1743-56. [PMID: 8886845 DOI: 10.1089/hum.1996.7.14-1743] [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] [Indexed: 02/02/2023] Open
Abstract
Development of a highly refined human factor IX (hFIX) expression vector system is critical for establishing a durable hemophilia B gene therapy. Here we report construction of a series of retroviral vectors and identification of an optimal basic structure and components for expressing hFIX in skeletal muscle cells. These vectors, which are derived from Moloney murine leukemia virus (MoMLV) with its enhancer sequence in the 3' long terminal repeat (LTR) deleted, contained internal hFIX expression units inserted in forward configuration without or with a viral vector intron sequence (pdL or pdLIn vector frame, respectively) or in inverted configuration without a viral vector intron sequence (pdLi frame). Internal expression units contained a hFIX cDNA or hFIX minigene (hIXm1 or hIXm2) derived from the hFIX cDNA by insertion of a shortened first intron sequence of the hFIX gene. Regardless of the promoter and vector frame used, both hIXm1 and hIXm2 gave 10- to 14-fold higher hFIX expression compared to those with hFIX cDNA. Internal hFIX transcriptional control units of these vectors were composed of various promoters linked with or without the muscle creatine kinase enhancer (Me) sequence. Promoters tested included those of alpha-actin (alpha A775), beta-actin (beta A280), cytochrome oxidase (CO1250 and CO650), myogenin (Mg1031 and Mg353), and Rous sarcoma virus (RSV). beta A200, which was derived from beta A280 by eliminating potential polyadenylation sites, was also tested. As extensively examined with the myogenin promoter, presence of one or multiple copies of Me in the vectors elevated the expression activity in myotubes by 4.5- to 19-fold over those without Me, but not significantly in myoblasts. Similar enhancements in expression activity with Me were also observed with other promoters, except those of RSV and CO. The latter two showed only modest enhancements in the presence of Me. As assayed with myotubes in culture, the general order of hFIX expression activity of various promoters with four copies of Me in the three different vector frames was beta A280 approximately beta A200 > Mg353 > Mg1031 approximately RSV approximately CO650 approximately alpha A775 > CO1250. One exception was that CO650 showed significantly less activity in pdLi-type vectors than in the pdLIn vectors. Based on the systematic analyses of various structural components, a group of pdLi vectors consisting of beta A200, two to four copies of Me, and hIXm2 was identified to have the optimal basic vector structure to be used in retrovirus for hFIX expression in differentiated skeletal muscle cells. The present studies provide the critical first step for establishing a highly refined hemophilia B gene therapy based on skeletal muscle-targeted hFIX gene transfer.
Collapse
Affiliation(s)
- J M Wang
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor 48109, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Yamashita T, Horio Y, Yamada M, Takahashi N, Kondo C, Kurachi Y. Competition between Mg2+ and spermine for a cloned IRK2 channel expressed in a human cell line. J Physiol 1996; 493 ( Pt 1):143-56. [PMID: 8735700 PMCID: PMC1158956 DOI: 10.1113/jphysiol.1996.sp021370] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
1. A cloned inwardly rectifying K+ channel, IRK2, was expressed in a human cell line, human embryonic kidney (HEK) 293T. Its electrophysiological properties were examined using the patch clamp technique in the whole-cell, cell-attached and inside-out patch configurations. 2. The cells transfected with IRK2 cDNA exhibited a K+ current which showed classical properties of inwardly rectifying K+ channels at both whole-cell and single-channel levels. 3. In the inside-out patch configuration, intracellular Mg2+ (Mg2+i blocked the outward currents in a voltage-dependent and virtually time-independent manner. Mg2+i (1-100 microM) caused a decrease in the unitary current amplitude of the IRK2 channel by inducing subconducting levels. 4. In the absence of Mg2+i, intracellular spermine blocked the outwardly flowing IRK2 currents in a voltage- and time-dependent manner. Spermine (1-100 nM) did not affect the unitary channel current amplitude but reduced the channel open probability. The spermine block showed a slower time and steeper voltage dependence than the Mg2+i++ block. 5. When both these blockers were present, Mg2+i apparently attenuated the inhibitory effect of spermine on the outwardly flowing IRK2 currents. This interaction was voltage and time dependent, and could be well explained by a model in which Mg2+i and spermine competitively bind to the channel with their individual first-order kinetics. This competition would induce time-dependent transits of the channel between the Mg2+i -and spermine-blocked states via a single open state, thereby preserving a certain size of persistent outward currents at depolarized potentials.
Collapse
Affiliation(s)
- T Yamashita
- Department of Pharmacology II, Faculty of Medicine, Osaka University, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Valentino TL, Lukasiewicz PD, Romano C. Immunocytochemical localization of polyamines in the tiger salamander retina. Brain Res 1996; 713:278-85. [PMID: 8725001 DOI: 10.1016/0006-8993(95)01558-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The polyamines spermine and spermidine are present in neural tissue, but their functions there are not well understood. Recent work suggests that the NMDA subtype of glutamate receptors, other glutamate receptor subtypes, and certain K(+)-channels, are neural targets for polyamines. To better understand the neuron-specific roles of polyamines, we have developed antibodies that interact with spermine and spermidine in aldehyde-fixed tissue and used these antibodies in immunocytochemical studies to determine the cellular localization of these polyamines in the tiger salamander retina. The affinity-purified, polyclonal antibodies were highly specific for spermine and spermidine, exhibiting < 1% cross reactivity with putrescine, and virtually no cross-reactivity with GABA, arginine, lysine, or glutaraldehyde. Polyamine labeling was most abundant in cells in the inner half of the inner nuclear layer and in the ganglion cell layer. Some cells in the outer half of the inner nuclear layer are labeled, and there was some labeling in both synaptic layers. Double-labeling experiments indicated (1) all GABAergic amacrine cells were polyamine-positive; and (2) all ganglion cells (identified by back-filling after microinjections of rhodamine in the optic nerve) were polyamine-positive. These results are consistent with a role for polyamines as modulators of NMDA receptor function and channel function in the inner retina.
Collapse
Affiliation(s)
- T L Valentino
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|