Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters











Publication year range
1.
Mult Scler Relat Disord ; 48: 102719, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33383365

ABSTRACT

Multiparametric flow cytometry (FC) of CSF allows one to easily estimate the percentage of lymphocyte subpopulations in CSF. We hypothesized that an increased ratio of B-lineage cells in CSF of MS patients, as assessed with FC, could be useful for diagnostics. We analyzed CSF of 137 patients (70 MS, 24 infectious/autoimmune neurologic disorders (INDs), and 43 non-infectious/autoimmune neurologic disorders (NINDs)), and showed that CSF plasmablasts of >0.1% had a sensitivity of 40% for MS and specificity of 92% when comparing MS and IND, while plasmablasts of >0.25% had sensitivity of 36%, and 100% specificity.


Subject(s)
Multiple Sclerosis , Nervous System Diseases , Blood Cell Count , Flow Cytometry , Humans , Nervous System Diseases/diagnosis , Plasma Cells
2.
Acta Neurol Scand ; 132(4): 278-83, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25819084

ABSTRACT

OBJECTIVE: The possibility of co-association between diabetes mellitus (DM) and chronic inflammatory demyelinating polyneuropathy (CIDP) has long been a focus of interest as well as of clinical significance. As CIDP is a potentially treatable condition, it is diagnosis in the context of DM is of great importance. However, diagnostic criteria to identify CIDP in patients with diabetes are not available. We propose a diagnostic tool that should help clinicians to decide what is the probability that a patient with diabetes might have CIDP. METHODS: We list several clinical, electrophysiological, and laboratory parameters that, when combined, have the power of discriminating an immune-mediated neuropathy in patients with DM. By summing the points assigned to each of these parameters, we define four levels of probability for a patient with diabetes to have CIDP. To analyze the validity of the diagnostic toll, we applied it in three different patient populations: (i) Patients with diabetes with peripheral neuropathy, (ii) Patients with CIDP without DM, and (iii) Patients with diabetes with CIDP. RESULTS: The scores of patients with diabetes without CIDP ranged from -7 to 2, while those of patients with DM-CIDP ranged from 2 to 20. The scores of non-diabetic patients with CIDP were similar to those of patients with DM-CIDP and ranged from 6 to 16. The mean score of patients with DM-CIDP was 9.083, while the score of patients with CIDP was 11.16 and that of patients with diabetic polyneuropathy was -3.59. CONCLUSIONS: These results show that this diagnostic tool is able to identify patients with diabetes with overlapping CIDP.


Subject(s)
Diabetic Neuropathies/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Aged , Female , Humans , Male , Middle Aged
3.
J Neurol Sci ; 344(1-2): 193-7, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25001515

ABSTRACT

Finglimod, a sphingosine 1-phosphate receptor modulator, is the first orally administered therapy approved for prophylaxis in multiple sclerosis (MS). Several reports in the last two years suggested that it might be associated with severe augmentation of disease activity upon initiation or discontinuation of therapy. We present an MS patient who developed a giant cavitating brain lesion under fingolimod and in whom cessation of therapy was associated with a very active course. Brain biopsy revealed the lesion to be due to an active demyelinating inflammatory process. With the current wave of immunosuppressive treatments for MS, there is a need to be vigilant to side effects and risks not identified in large multicenter trials, collect the data and set guidelines and precautions for present and future medications.


Subject(s)
Brain Injuries/chemically induced , Encephalitis/chemically induced , Immunosuppressive Agents/adverse effects , Multiple Sclerosis/drug therapy , Occipital Lobe/pathology , Propylene Glycols/adverse effects , Sphingosine/analogs & derivatives , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Female , Fingolimod Hydrochloride , Humans , Inflammation/chemically induced , Magnetic Resonance Imaging , Sphingosine/adverse effects
4.
J Neurol Sci ; 338(1-2): 39-42, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24267740

ABSTRACT

OBJECTIVES: There is insufficient data on the efficacy of intravenous immunoglobulins (IVIg) as maintenance treatment in myasthenia gravis (MG). We therefore examined response to maintenance IVIg therapy in a cohort of MG patients. METHODS: We reviewed all MG patient files treated with IVIg in our neuro-immunology clinic from 1/1995 to 9/2012. Patients treated with maintenance IVIg for a minimum of one year were separately analyzed. Disease severity was evaluated according to the Myasthenia Gravis Foundation of America clinical classification. RESULTS: IVIg was considered for maintenance therapy in 52 MG patients who had not responded to pyridostigmine, prednisone, azathioprine or combinations of these drugs. Fifteen patients did not improve with initial IVIg while thirty seven patients had a beneficial response and were treated with maintenance IVIg for an average of 5.9 years (range 1-17 years). Twenty two (59%) patients were female with an average age onset of disease 44.8 years. Thirty three were seropositive for acetylcholine receptor antibody and 13 had previous thymectomy. Twenty three and 14 patients achieved mild or moderate improvement respectively in disease activity while on IVIg therapy but none achieved full remission. Beneficial response was associated with older age, bulbar presentation, seropositivity and a higher antibody titer and less with ocular presentation. IVIg enabled reduction of other treatments including pyridostigmine, prednisone and azathioprine. CONCLUSION: In this retrospective study on a relative small cohort of MG patients maintenance IVIg therapy was successful in reducing symptoms of MG but seems to be ineffective in inducing full remission or reducing disease activity. IVIg should be regarded only as symptomatic therapy in MG.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , Myasthenia Gravis/immunology , Myasthenia Gravis/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies/blood , Child , Cohort Studies , Female , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Receptors, Cholinergic/immunology , Treatment Outcome , Young Adult
5.
Diabetologia ; 55(6): 1709-20, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22411134

ABSTRACT

AIMS/HYPOTHESIS: It is thought that the voltage-dependent potassium channel subunit Kv2.1 (Kv2.1) regulates insulin secretion by controlling beta cell electrical excitability. However, this role of Kv2.1 in human insulin secretion has been questioned. Interestingly, Kv2.1 can also regulate exocytosis through direct interaction of its C-terminus with the soluble NSF attachment receptor (SNARE) protein, syntaxin 1A. We hypothesised that this interaction mediates insulin secretion independently of Kv2.1 electrical function. METHODS: Wild-type Kv2.1 or mutants lacking electrical function and syntaxin 1A binding were studied in rodent and human beta cells, and in INS-1 cells. Small intracellular fragments of the channel were used to disrupt native Kv2.1-syntaxin 1A complexes. Single-cell exocytosis and ion channel currents were monitored by patch-clamp electrophysiology. Interaction between Kv2.1, syntaxin 1A and other SNARE proteins was probed by immunoprecipitation. Whole-islet Ca(2+)-responses were monitored by ratiometric Fura red fluorescence and insulin secretion was measured. RESULTS: Upregulation of Kv2.1 directly augmented beta cell exocytosis. This happened independently of channel electrical function, but was dependent on the Kv2.1 C-terminal syntaxin 1A-binding domain. Intracellular fragments of the Kv2.1 C-terminus disrupted native Kv2.1-syntaxin 1A interaction and impaired glucose-stimulated insulin secretion. This was not due to altered ion channel activity or impaired Ca(2+)-responses to glucose, but to reduced SNARE complex formation and Ca(2+)-dependent exocytosis. CONCLUSIONS/INTERPRETATION: Direct interaction between syntaxin 1A and the Kv2.1 C-terminus is required for efficient insulin exocytosis and glucose-stimulated insulin secretion. This demonstrates that native Kv2.1-syntaxin 1A interaction plays a key role in human insulin secretion, which is separate from the channel's electrical function.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Shab Potassium Channels/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Electrophysiology , Humans , Immunoblotting , Immunoprecipitation , Insulin Secretion , Mice , Protein Binding , Rats , Shab Potassium Channels/genetics , Syntaxin 1/metabolism
7.
J Neurosci ; 21(6): 1964-74, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11245681

ABSTRACT

Presynaptic voltage-gated K(+) (Kv) channels play a physiological role in the regulation of transmitter release by virtue of their ability to shape presynaptic action potentials. However, the possibility of a direct interaction of these channels with the exocytotic apparatus has never been examined. We report the existence of a physical interaction in brain synaptosomes between Kvalpha1.1 and Kvbeta subunits with syntaxin 1A, occurring, at least partially, within the context of a macromolecular complex containing syntaxin, synaptotagmin, and SNAP-25. The interaction was altered after stimulation of neurotransmitter release. The interaction with syntaxin was further characterized in Xenopus oocytes by both overexpression and antisense knock-down of syntaxin. Direct physical interaction of syntaxin with the channel protein resulted in an increase in the extent of fast inactivation of the Kv1.1/Kvbeta1.1 channel. Syntaxin also affected the channel amplitude in a biphasic manner, depending on its concentration. At low syntaxin concentrations there was a significant increase in amplitudes, with no detectable change in cell-surface channel expression. At higher concentrations, however, the amplitudes decreased, probably because of a concomitant decrease in cell-surface channel expression, consistent with the role of syntaxin in regulation of vesicle trafficking. The observed physical and functional interactions between syntaxin 1A and a Kv channel may play a role in synaptic efficacy and neuronal excitability.


Subject(s)
Antigens, Surface/metabolism , Brain/metabolism , Calcium-Binding Proteins , Ion Channel Gating/physiology , Membrane Proteins , Nerve Tissue Proteins/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , Antigens, Surface/genetics , Antigens, Surface/pharmacology , Cells, Cultured , Exocytosis/physiology , Ion Channel Gating/drug effects , Kv1.1 Potassium Channel , Membrane Glycoproteins/metabolism , Microinjections , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurotransmitter Agents/metabolism , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Protein Binding , Protein Subunits , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synaptosomal-Associated Protein 25 , Synaptosomes/metabolism , Synaptotagmins , Syntaxin 1 , Xenopus
8.
Pflugers Arch ; 440(4): 627-33, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10958347

ABSTRACT

We describe the preparation of a Xenopus oocyte plasma membrane patch attached to a cover-slip with its intracellular face exposed to the bath solution. The proteins attached to the plasma membrane were visualized by confocal microscopy after fluorescence labelling. Since cortical microfilament elements were detected in these plasma membrane preparations we termed the patches plasma membrane-cortex patches. The way these patches are formed and the low concentration of proteins needed for cytochemical detection make the membrane-cortex patches similar to electrophysiological membrane patches and therefore allow the cytochemical study of ion channels to be correlated with electrophysiological experiments. Furthermore, the described patch is similar to manually isolated plasma membranes used for biochemical analysis by sodium dodecylsulphate-polyacrylamide gel electrophoresis (SDS-PAGE). Cytochemical analysis of membrane-cortex patches also enables the detection of the two-dimensional pattern of organization of membrane proteins (clustered or non-clustered forms). In addition, patch preparations enable cytochemical study of the relative localization of membrane proteins. The methodology enables integration of electrophysiological, biochemical and cytochemical studies of ion channels, giving a comprehensive perspective on ion channel function.


Subject(s)
Cell Membrane/chemistry , Membrane Proteins/analysis , Oocytes/ultrastructure , Potassium Channels, Inwardly Rectifying , Xenopus laevis , Actins/analysis , Animals , Antigens, Surface/analysis , Calcium Channels/analysis , Cytochalasin D/pharmacology , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Female , Fluorescein-5-isothiocyanate , Fluorescent Dyes , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Microscopy, Fluorescence , Nerve Tissue Proteins/analysis , Phalloidine , Potassium Channels/analysis , Potassium Channels/genetics , RNA, Messenger/analysis , Syntaxin 1
9.
J Neurosci ; 19(20): 8730-9, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516292

ABSTRACT

Several scorpion toxins have been shown to exert their neurotoxic effects by a direct interaction with voltage-dependent sodium channels. Both classical scorpion alpha-toxins such as Lqh II from Leiurus quiquestratus hebraeus and alpha-like toxins as toxin III from the same scorpion (Lqh III) competitively interact for binding on receptor site 3 of insect sodium channels. Conversely, Lqh III, which is highly toxic in mammalian brain, reveals no specific binding to sodium channels of rat brain synaptosomes and displaces the binding of Lqh II only at high concentration. The contrast between the low-affinity interaction and the high toxicity of Lqh III indicates that Lqh III binding sites distinct from those present in synaptosomes must exist in the brain. In agreement, electrophysiological experiments performed on acute rat hippocampal slices revealed that Lqh III strongly affects the inactivation of voltage-gated sodium channels recorded either in current or voltage clamp, whereas Lqh II had weak, or no, effects. In contrast, Lqh III had no effect on cultured embryonic chick central neurons and on sodium channels from rat brain IIA and beta1 subunits reconstituted in Xenopus oocytes, whereas sea anemone toxin ATXII and Lqh II were very active. These data indicate that the alpha-like toxin Lqh III displays a surprising subtype specificity, reveals the presence of a new, distinct sodium channel insensitive to Lqh II, and highlights the differences in distribution of channel expression in the CNS. This toxin may constitute a valuable tool for the investigation of mammalian brain function.


Subject(s)
Brain/metabolism , Neurons/metabolism , Peptides , Sodium Channels/metabolism , Animals , Brain/cytology , Chick Embryo , Cockroaches/drug effects , Female , Intercellular Signaling Peptides and Proteins , Oocytes/metabolism , Peptides/metabolism , Peptides/pharmacology , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Sodium Channels/drug effects , Substrate Specificity , Tissue Distribution , Xenopus laevis
10.
EMBO J ; 18(5): 1245-56, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10064591

ABSTRACT

Modulation of A-type voltage-gated K+ channels can produce plastic changes in neuronal signaling. It was shown that the delayed-rectifier Kv1.1 channel can be converted to A-type upon association with Kvbeta1.1 subunits; the conversion is only partial and is modulated by phosphorylation and microfilaments. Here we show that, in Xenopus oocytes, expression of Gbeta1gamma2 subunits concomitantly with the channel (composed of Kv1.1 and Kvbeta1.1 subunits), but not after the channel's expression in the plasma membrane, increases the extent of conversion to A-type. Conversely, scavenging endogenous Gbetagamma by co-expression of the C-terminal fragment of the beta-adrenergic receptor kinase reduces the extent of conversion to A-type. The effect of Gbetagamma co-expression is occluded by treatment with dihydrocytochalasin B, a microfilament-disrupting agent shown previously by us to enhance the extent of conversion to A-type, and by overexpression of Kvbeta1.1. Gbeta1gamma2 subunits interact directly with GST fusion fragments of Kv1.1 and Kvbeta1.1. Co-expression of Gbeta1gamma2 causes co-immunoprecipitation with Kv1.1 of more Kvbeta1.1 subunits. Thus, we suggest that Gbeta1gamma2 directly affects the interaction between Kv1.1 and Kvbeta1.1 during channel assembly which, in turn, disrupts the ability of the channel to interact with microfilaments, resulting in an increased extent of A-type conversion.


Subject(s)
Brain/metabolism , GTP-Binding Proteins/metabolism , Potassium Channels/genetics , Actin Cytoskeleton/metabolism , Animals , Cytochalasin B/analogs & derivatives , Cytochalasin B/pharmacology , Electrophysiology , Gene Expression Regulation/genetics , Ion Channel Gating/physiology , Microinjections , Oocytes/metabolism , Phosphorylation , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Binding/genetics , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics , Xenopus laevis
11.
Pflugers Arch ; 439(1-2): 18-26, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10650996

ABSTRACT

Modulation of fast-inactivating voltage-gated K+ channels can produce plastic changes in neuronal signaling. Previously, we showed that the voltage-dependent K+ channel composed of brain Kv1.1 and Kvbeta1.1 subunits (alpha(beta) channel) gives rise to a current that has a fast-inactivating and a sustained component; the proportion of the fast-inactivating component could be decreased by dephosphorylation of a basally phosphorylated Ser-446 on the alpha subunit. To account for our results we suggested a model that assumes a bimodal gating of the alpha(beta) channel. In this study, using single-channel analysis, we confirm this model. Two modes of gating were identified: (1) an inactivating mode characterized by low open probability and single openings early in the voltage step, and (2) a non-inactivating gating mode with bursts of openings. These two modes were non-randomly distributed, with spontaneous shifts between them. Each mode is characterized by a different set of open time constants (tau) and mean open times (t(0)). The non-inactivating mode is similar to the gating mode of a homomultimeric alpha channel. The phosphorylation-deficient alphaS446Abeta channel has the same two gating modes. Furthermore, alkaline phosphatase promoted the transition to the non-inactivating mode. This is the first report of modal behavior of a fast-inactivating K+ channel; furthermore, it substantiates the notion that direct phosphorylation is one mechanism that regulates the equilibrium between the two modes and thereby regulates the extent of macroscopic fast inactivation of a brain K+ channel.


Subject(s)
Ion Channel Gating/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Algorithms , Alkaline Phosphatase/metabolism , Animals , Kv1.1 Potassium Channel , Membrane Potentials/physiology , Models, Biological , Oocytes/metabolism , Patch-Clamp Techniques , Phosphorylation , Potassium Channels/metabolism , Xenopus laevis
12.
Pflugers Arch ; 436(1): 56-68, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9560447

ABSTRACT

The G-protein-activated K+ channels of the GIRK (Kir 3) family are activated by Gbetagamma subunits of heterotrimeric Gi/Go proteins. Atrial GIRK currents evoked by acetylcholine (ACh)1 via muscarinic m2 receptors (m2R) display prominent desensitization. We studied desensitization of basal and ACh-evoked whole-cell GIRK currents in Xenopus oocytes. In the absence of receptor and/or agonist, the basal GIRK activity showed inactivation which was prominent when the preparation was bathed in a low-Na+, high-K+ extracellular solution (96 mM [K+]out and 2 mM [Na+]out) but did not occur in a normal physiological solution. Ion substitution experiments showed that this basal, agonist-independent inactivation was caused by the decrease in [Na+]out rather than by the increased [K+]out. We hypothesize that it reflects a depletion of intracellular Na+. ACh-evoked GIRK currents desensitized faster than the basal ones. The agonist-induced desensitization was present when the preparation was bathed in all solutions tested, independently of [Na+]out. A protein kinase C (PKC) activator inhibited the GIRK currents both in high and low [Na+]out, apparently mimicking agonist-induced desensitization; however, a potent serine/threonine protein kinase blocker, staurosporine, blocked only a minor part of desensitization. We conclude that basal inactivation and agonist-induced desensitization are separate processes, the former caused by changes in Na+ concentrations, and the latter by unknown factor(s) with only a minor contribution of PKC.


Subject(s)
Acetylcholine/pharmacology , GTP-Binding Proteins/physiology , Oocytes/physiology , Potassium Channels/physiology , Animals , Drug Tolerance , Electric Conductivity , Enzyme Activation/drug effects , Female , Potassium/administration & dosage , Potassium Channels/drug effects , Protein Kinase C/metabolism , Receptor, Muscarinic M2 , Receptors, Muscarinic/physiology , Sodium/administration & dosage , Sodium/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
13.
J Biol Chem ; 273(11): 6495-502, 1998 Mar 13.
Article in English | MEDLINE | ID: mdl-9497384

ABSTRACT

Various brain K+ channels, which may normally exist as complexes of alpha (pore-forming) and beta (auxiliary) subunits, were subjected to regulation by metabotropic glutamate receptors. Kv1.1/Kvbeta1.1 is a voltage-dependent K+ channel composed of alpha and beta proteins that are widely expressed in the brain. Expression of this channel in Xenopus oocytes resulted in a current that had fast inactivating and noninactivating components. Previously we showed that basal and protein kinase A-induced phosphorylation of the alpha subunit at Ser-446 decreases the fraction of the noninactivating component. In this study we investigated the effect of protein kinase C (PKC) on the channel. We showed that a PKC-activating phorbol ester (phorbol 12-myristate 13-acetate (PMA)) increased the noninactivating fraction via activation of a PKC subtype that was inhibited by staurosporine and bisindolylmaleimide but not by calphostin C. However, it was not a PKC-induced phosphorylation but rather a dephosphorylation that mediated the effect. PMA reduced the basal phosphorylation of Ser-446 significantly in plasma membrane channels and failed to affect the inactivation of channels having an alpha subunit that was mutated at Ser-446. Also, the activation of coexpressed mGluR1a known to activate phospholipase C mimicked the effect of PMA on the inactivation via induction of dephosphorylation at Ser-446. Thus, this study identified a potential neuronal pathway initiated by activation of metabotropic glutamate receptor 1a coupled to a signaling cascade that possibly utilized PKC to induce dephosphorylation and thereby to decrease the extent of inactivation of a K+ channel.


Subject(s)
Ion Channel Gating/drug effects , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Protein Kinase C/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Electric Conductivity , Electrophysiology/methods , Kv1.1 Potassium Channel , Molecular Sequence Data , Oocytes , Patch-Clamp Techniques , Phosphorylation , Protein Binding , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
14.
J Biol Chem ; 272(22): 14021-4, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9162020

ABSTRACT

Kv1.1/Kvbeta1.1 (alphabeta) K+ channel expressed in Xenopus oocytes was shown to have a fast inactivating current component. The fraction of this component (extent of inactivation) is increased by microfilament disruption induced by cytochalasins or by phosphorylation of the alpha subunit at Ser-446, which impairs the interaction of the channel with microfilaments. The relevant sites of interaction on the channel molecules have not been identified. Using a phosphorylation-deficient mutant of alpha, S446A, to ensure maximal basal interaction of the channel with the cytoskeleton, we show that one relevant site is the end of the C terminus of alpha. Truncation of the last six amino acids resulted in alphabeta channels with an extent of inactivation up to 2.5-fold larger and its further enhancement by cytochalasins being reduced 2-fold. The wild-type channels exhibited strong inactivation, which could not be markedly increased either by cytochalasins or by the C-terminal mutations, indicating that the interaction of the wild-type channels with microfilaments was minimal to begin with, presumably because of extensive basal phosphorylation. Since the C-terminal end of Kv1.1 was shown to participate in channel clustering via an interaction with members of the PSD-95 family of proteins, we propose that a similar interaction with an endogenous protein takes place, contributing to channel connection to the oocyte cytoskeleton. This is the first report to assign a modulatory role to such an interaction: together with the state of phosphorylation of the channel, it regulates the extent of inactivation conferred by the beta subunit.


Subject(s)
Cytoskeleton/physiology , Potassium Channels/physiology , Amino Acid Sequence , Animals , Female , Molecular Sequence Data , Oocytes/metabolism , Phosphorylation , Xenopus
15.
J Biol Chem ; 271(46): 29321-8, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8910593

ABSTRACT

Voltage-gated K+ channels isolated from mammalian brain are composed of alpha and beta subunits. Interaction between coexpressed Kv1.1 (alpha) and Kvbeta1.1 (beta) subunits confers rapid inactivation on the delayed rectifier-type current that is observed when alpha subunits are expressed alone. Integrating electrophysiological and biochemical analyses, we show that the inactivation of the alphabeta current is not complete even when alpha is saturated with beta, and the alphabeta current has an inherent sustained component, indistinguishable from a pure alpha current. We further show that basal and protein kinase A-induced phosphorylations at Ser-446 of the alpha protein increase the extent, but not the rate, of inactivation of the alphabeta channel, without affecting the association between alpha and beta. In addition, the extent of inactivation is increased by agents that lead to microfilament depolymerization. The effects of phosphorylation and of microfilament depolymerization are not additive. Taken together, we suggest that phosphorylation, via a mechanism that involves the interaction of the alphabeta channel with microfilaments, enhances the extent of inactivation of the channel. Furthermore, phosphorylation at Ser-446 also increases current amplitudes of the alphabeta channel as was shown before for the alpha channel. Thus, phosphorylation enhances in concert inactivation and current amplitudes, thereby leading to a substantial increase in A-type activity.


Subject(s)
Cytoskeleton/metabolism , Potassium Channels/metabolism , Animals , Phosphorylation , Potassium Channel Blockers , Potassium Channels/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenopus
16.
FEBS Lett ; 384(2): 189-92, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8612821

ABSTRACT

The well-characterized enhancement of the cardiac Ca2+ L-type current by protein kinase A (PKA) is not observed when the corresponding channel is expressed in Xenopus oocytes, possibly because it is fully phosphorylated in the basal state. However, the activity of the expressed channel is reduced by PKA inhibitors. Using this paradigm as an assay to search for PKA sites relevant to channel modulation, we have found that mutation of serine 1928 of the alpha 1C subunit to alanine abolishes the modulation of the expressed channel by PKA inhibitors. This effect was independent of the presence of the beta subunit. Phosphorylation of serine 1928 of alpha 1C may mediate the modulatory effect of PKA on the cardiac voltage-dependent ca2+ channel.


Subject(s)
Calcium Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Muscle Proteins/metabolism , Myocardium/chemistry , Protein Processing, Post-Translational , Sulfonamides , Animals , Base Sequence , Calcium Channels/chemistry , Calcium Channels, L-Type , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ion Channel Gating/drug effects , Isoquinolines/pharmacology , Molecular Sequence Data , Muscle Proteins/chemistry , Mutagenesis, Site-Directed , Oocytes , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rabbits , Recombinant Fusion Proteins/metabolism , Serine/chemistry , Thionucleotides/pharmacology , Xenopus laevis
17.
FEBS Lett ; 381(1-2): 71-6, 1996 Feb 26.
Article in English | MEDLINE | ID: mdl-8641443

ABSTRACT

The modulation by protein kinase C (PKC) of the RCK1 K+ channel was investigated in Xenopus oocytes by integration of two-electrode voltage clamp, site-directed mutagenesis and SDS-PAGE analysis techniques. Upon application of beta-phorbol 12-myristate 13-acetate (PMA) the current was inhibited by 50-90%. No changes in the voltage sensitivity of the channel, changes in membrane surface area or selective elimination of RCK1 protein from the plasma membrane could be detected. The inhibition was mimicked by 1-oleoyl-2-acetyl-rac-glycerol (OAG) but not by alphaPMA, and was blocked by staurosporine and calphostin C. Upon deletion of most of the N-terminus a preceding enhancement of about 40% of the current was prominent in response to PKC activation. Its physiological significance is discussed. The N-terminus deletion eliminated 50% of the inhibition. However, phosphorylation of none of the ten classical PKC phosphorylation sites on the channel molecule could account, by itself or in combination with others, for the inhibition. Thus, our results show that PKC activation can modulate the channel conductance in a bimodal fashion. The N-terminus is involved in the inhibition, however, not via its direct phosphorylation.


Subject(s)
Brain/metabolism , Oocytes/physiology , Potassium Channels/physiology , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Alkaloids/pharmacology , Animals , Base Sequence , Cell Membrane/drug effects , Cell Membrane/physiology , DNA Primers , Diglycerides/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Kinetics , Membrane Potentials/drug effects , Models, Structural , Molecular Sequence Data , Mutagenesis, Site-Directed , Naphthalenes/pharmacology , Oocytes/drug effects , Patch-Clamp Techniques , Potassium Channels/biosynthesis , Potassium Channels/chemistry , Protein Structure, Secondary , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Staurosporine , Time Factors , Xenopus laevis
18.
J Mol Neurosci ; 7(4): 269-76, 1996.
Article in English | MEDLINE | ID: mdl-8968948

ABSTRACT

On deletion of the N-terminus of RCK1 K+ channel, acute modulation of the channel by cAMP-elevating treatments is revealed. This modulation is studied in Xenopus oocytes using two-electrode voltage-clamp, site-directed mutagenesis, and SDS-PAGE analyses. Treatments by Sp-8-Br-cAMPS, a membrane-permeant cAMP analog, and by isoproterenol, a beta 1-adrenergic receptor (beta 1R) agonist, both increased the current amplitudes with no effect on the voltage dependency of activation. The effect of isoproterenol was blocked by coexpression of either G alpha S or G alpha i3 proteins. The channel protein is phosphorylated on the Sp-8-Br-cAMPS treatment at Ser446; however, a phosphorylation-deficient variant in which this site has been altered is still modulated by Sp-8-Br-cAMPS and isoproterenol. Expression of the full-length channel with Kv beta 1.1 auxiliary subunit renders the channel at the same modulation as that of the truncated one. Taken together, the RCK1 channel can be acutely modulated by cAMP and beta 1R activation possibly through protein kinase A (PKA) activation, but not through direct channel phosphorylation; the involvement of the N-terminus in this modulation is discussed.


Subject(s)
Cyclic AMP/analogs & derivatives , Cyclic AMP/physiology , Isoproterenol/pharmacology , Potassium Channels/physiology , Receptors, Adrenergic, beta-1/physiology , Thionucleotides/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/metabolism , Macromolecular Substances , Membrane Potentials/drug effects , Mutagenesis, Site-Directed , Oocytes/drug effects , Oocytes/physiology , Potassium Channels/biosynthesis , Potassium Channels/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Serine , Xenopus laevis
19.
J Biol Chem ; 270(24): 14611-8, 1995 Jun 16.
Article in English | MEDLINE | ID: mdl-7782324

ABSTRACT

We have recently shown that the rat brain Kv1.1 (RCK1) voltage-gated K+ channel is partially phosphorylated in its basal state in Xenopus oocytes and can be further phosphorylated upon treatment for a short time with a cAMP analog (Ivanina, T., Perts, T., Thornhill, W. B., Levin, G., Dascal, N., and Lotan, I. (1994) Biochemistry 33, 8786-8792). In this study, we show, by two-electrode voltage clamp analysis, that whereas treatments for a short time with various cAMP analogs do not affect the channel function, prolonged treatment with 8-bromoadenosine 3',5'-cyclic monophosphorothioate ((Sp)-8-Br-cAMPS), a membrane-permeant cAMP analog, enhances the current amplitude. It also enhances the current amplitude through a mutant channel that cannot be phosphorylated by protein kinase A activation. The enhancement is inhibited in the presence of (Rp)-8-Br-cAMPS, a membrane-permeant protein kinase A inhibitor. Concomitant SDS-polyacrylamide gel electrophoresis analysis reveals that this treatment not only brings about phosphorylation of the wild-type channel, but also increases the amounts of both wild-type and mutant channel proteins; the latter effect can be inhibited by cycloheximide, a protein synthesis inhibitor. In the presence of cycloheximide, the (Sp)-8-Br-cAMPS treatment enhances only the wild-type current amplitudes and induces accumulation of wild-type channels in the plasma membrane of the oocyte. In summary, prolonged treatment with (Sp)-8-Br-cAMPS regulates RCK1 function via two pathways, a pathway leading to enhanced channel synthesis and a pathway involving channel phosphorylation that directs channels to the plasma membrane.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Kv1.1 Potassium Channel , Molecular Sequence Data , Mutation , Oligodeoxyribonucleotides , Phosphorylation , Potassium Channels/genetics , Potassium Channels/metabolism , Xenopus
20.
Biochemistry ; 33(29): 8786-92, 1994 Jul 26.
Article in English | MEDLINE | ID: mdl-8038169

ABSTRACT

Phosphorylation-mediated regulation of voltage-gated K+ channels has been implicated in numerous electrophysiological studies; however, complementary biochemical studies have so far been hampered by the failure to isolate and characterize any K+ channel proteins of distinct molecular identity. We used the Xenopus oocyte expression system to study the biosynthesis and phosphorylation by protein kinase A (PKA) of rat brain RCK1 (Kv1.1) K+ channel protein. RCK1 protein was isolated by immunoprecipitation from oocytes injected with RCK1 cRNA and analyzed by SDS-polyacrylamide gel electrophoresis (SDS-PAGE). The channel protein was expressed in the form of several polypeptides. The 57-kDa polypeptide, usually the major constituent, resided both in the cytosol and in the plasma membrane. Its levels were correlated with RCK1 current amplitudes (IRCK1) and upon incubation of the cRNA-injected oocytes with tunicamycin, its molecular weight was decreased and at the same time IRCK1 was reduced. These results suggest that the membranal 57-kDa polypeptides represent functional channels that are N-glycosylated. Furthermore, a study of the phosphorylation of the RCK1 polypeptides revealed that the 57-kDa polypeptide was specifically targeted for phosphorylation by PKA. It could be phosphorylated in vitro by the catalytic subunit of PKA (PKA-CS). In its native state in intact oocytes, the 57-kDa polypeptide was partially phosphorylated and could be further phosphorylated in vivo by addition of a membrane-permeant cAMP analog. Site-directed mutagenesis demonstrated that phosphorylation of a single site on the C-terminus of the channel molecule fully accounts for these phosphorylations.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Potassium Channels/metabolism , Animals , Base Sequence , Cell Membrane/enzymology , Cyclic AMP/metabolism , DNA Primers/chemistry , Ion Channel Gating , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes , Phosphorylation , Recombinant Proteins , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL