Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 620, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32001697

ABSTRACT

Sleeping sickness is a fatal disease caused by the protozoan parasite Trypanosoma brucei (Tb). Inosine-5'-monophosphate dehydrogenase (IMPDH) has been proposed as a potential drug target, since it maintains the balance between guanylate deoxynucleotide and ribonucleotide levels that is pivotal for the parasite. Here we report the structure of TbIMPDH at room temperature utilizing free-electron laser radiation on crystals grown in living insect cells. The 2.80 Å resolution structure reveals the presence of ATP and GMP at the canonical sites of the Bateman domains, the latter in a so far unknown coordination mode. Consistent with previously reported IMPDH complexes harboring guanosine nucleotides at the second canonical site, TbIMPDH forms a compact oligomer structure, supporting a nucleotide-controlled conformational switch that allosterically modulates the catalytic activity. The oligomeric TbIMPDH structure we present here reveals the potential of in cellulo crystallization to identify genuine allosteric co-factors from a natural reservoir of specific compounds.


Subject(s)
Coenzymes/chemistry , Crystallization , IMP Dehydrogenase/chemistry , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Cloning, Molecular , Guanosine Monophosphate , Models, Molecular , Protein Conformation , Sf9 Cells , Trypanosoma brucei brucei/genetics
2.
Struct Dyn ; 2(4): 041712, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26798811

ABSTRACT

X-ray crystallography requires sufficiently large crystals to obtain structural insights at atomic resolution, routinely obtained in vitro by time-consuming screening. Recently, successful data collection was reported from protein microcrystals grown within living cells using highly brilliant free-electron laser and third-generation synchrotron radiation. Here, we analyzed in vivo crystal growth of firefly luciferase and Green Fluorescent Protein-tagged reovirus µNS by live-cell imaging, showing that dimensions of living cells did not limit crystal size. The crystallization process is highly dynamic and occurs in different cellular compartments. In vivo protein crystallization offers exciting new possibilities for proteins that do not form crystals in vitro.

4.
Orthopade ; 37(8): 783-7, 2008 Aug.
Article in German | MEDLINE | ID: mdl-18629468

ABSTRACT

BACKGROUND: This retrospective study was performed to investigate the clinical and radiological results after STAR total ankle replacement. MATERIAL AND METHODS: Between January 2000 and September 2004, 49 patients with an average age of 62.5 years underwent total ankle replacement with the STAR prosthesis. At an average follow-up of 30.4 months, 48 patients were examined clinically and radiologically. The Kofoed ankle score and the patients' subjective satisfaction were evaluated. RESULTS: The operation improved the Kofoed ankle score significantly, from 28 to 86 points, 90% of the patients were satisfied with the results. The revision rate was 10%. CONCLUSION: The early results after implantation of the STAR ankle prosthesis are encouraging. With correct indication, a high rate of pain reduction and patient satisfaction can be achieved. The long-term benefit of this procedure has yet to be determined.


Subject(s)
Ankle Joint/surgery , Joint Prosthesis , Osteoarthritis/surgery , Vitallium , Adult , Aged , Ankle Joint/diagnostic imaging , Female , Humans , Male , Middle Aged , Osteoarthritis/diagnostic imaging , Patient Satisfaction , Postoperative Care , Postoperative Complications/diagnostic imaging , Prosthesis Design , Radiography , Range of Motion, Articular , Reoperation
5.
J Membr Biol ; 205(3): 175-84, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16362505

ABSTRACT

Ion channels have a critical role in cell proliferation and it is well documented that channel blockers can inhibit the growth of cancer cells. The concept of ion channels as therapeutic targets or prognostic biomarkers attracts increasing interest, but the lack of potent and selective channel modulators has hampered a critical verification for many years. Today, the knowledge of human ion channel genes is almost complete and molecular correlates for many native currents have already been identified. This information triggered a wave of experimental results, identifying individual ion channels with relevance for specific cancer types. The current pattern of cancer-related ion channels is not arbitrary, but can be reduced to few members from each ion channel family. This review aims to provide an overview of the molecularly identified ion channels that might be relevant for the most common human cancer types. Possible applications of these candidates for a targeted cancer therapy or for clinical diagnosis are discussed.


Subject(s)
Ion Channels , Neoplasms/diagnosis , Neoplasms/drug therapy , Biomarkers , Breast Neoplasms/physiopathology , Calcium Channels/physiology , Colorectal Neoplasms/physiopathology , Female , Humans , Ion Channels/drug effects , Ion Channels/physiology , Lymphoma, Non-Hodgkin/physiopathology , Male , Melanoma/physiopathology , Prognosis , Prostatic Neoplasms/physiopathology , TRPM Cation Channels/physiology , Urinary Bladder Neoplasms/physiopathology
6.
J Membr Biol ; 188(2): 137-49, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12172639

ABSTRACT

Human IGR1 cells are a model for malignant melanoma. Since progression through the cell cycle is accompanied by transient cell hyperpolarization, we studied the properties of potassium and chloride ion channels and their impact on cell growth. The major potassium current components were mediated by outward rectifying ether à go-go (hEAG) channels and Ca2+-activated channels (KCa) of the IK/SK type. The major chloride channel component was activated by osmotic cell swelling (Clvol). To infer about the contribution of these channels to proliferation, specific inhibitors are required. Since there is no specific blocker for hEAG available, we used the tricyclic antidepressant imipramine, which blocked all channels mentioned, in combination with blockers for KCa (charybdotoxin) and Clvol (DIDS and pamoic acid). Incubation of IGR1 cells for 48 hr in 10-15 mM imipramine reduced DNA synthesis and metabolism without significant effects on apoptosis. hEAG channels were most sensitive to imipramine (IC50: 3.4 microM at +50 mV), followed by KCa (13.8 microM at +50 mV) and Clvol (12 microM at -100 mV), indicating that hEAG expression may be of importance for proliferation of melanoma cells. The contribution of KCa channels could be excluded, as 500 nM charybdotoxin, which completely blocked KCa, had no effect on proliferation. The impact of Clvol also seems to be minor, because 500 microM pamoic acid, which completely blocked Clvol, did not affect proliferation either.


Subject(s)
Imipramine/pharmacology , Melanoma, Experimental/pathology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels/drug effects , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Division/drug effects , Charybdotoxin/pharmacology , Chloride Channels/drug effects , Chloride Channels/physiology , DNA , Humans , Ion Channel Gating/physiology , Ion Channels , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Naphthols/pharmacology , Patch-Clamp Techniques , Potassium Channels/physiology , Potassium Channels, Calcium-Activated/physiology , Tumor Cells, Cultured
7.
Biochem J ; 360(Pt 3): 691-8, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11736661

ABSTRACT

Type-I phosphoinositide 3-kinases (PI3Ks) were characterized as a group of intracellular signalling proteins expressing both protein and lipid kinase activities. Recent studies implicate PI3Ks as mediators of oocyte maturation, but the molecular mechanisms are poorly defined. Here we used the Xenopus oocyte expression system as a model to investigate a possible contribution of the gamma-isoform of PI3K (PI3Kgamma) in the different pathways leading to cell-cycle progression by monitoring the time course of germinal vesicle breakdown (GVBD). Expression of a constitutive active PI3Kgamma (PI3Kgamma-CAAX) induced GVBD and increased the levels of phosphorylated Akt/protein kinase B and mitogen-activated protein kinase (MAPK). Furthermore, PI3Kgamma-CAAX accelerated progesterone-induced GVBD, but had no effect on GVBD induced by insulin. The effects of PI3Kgamma-CAAX could be suppressed by pre-incubation of the oocytes with LY294002, PD98059 or roscovitine, inhibitors of PI3K, MEK (MAPK/extracellular-signal-regulated protein kinase kinase) and cdc2/cyclin B kinase, respectively. Mutants of PI3Kgamma-CAAX, in which either lipid kinase or both lipid and protein kinase activities were altered or eliminated, did not induce significant GVBD. Our data demonstrate that expression of PI3Kgamma in Xenopus oocytes accelerates their progesterone-induced maturation and that lipid kinase activity is required to induce this effect.


Subject(s)
Oocytes/physiology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Germinal Center/drug effects , Germinal Center/physiology , In Vitro Techniques , Insulin/pharmacology , Kinetics , Mitogen-Activated Protein Kinases/isolation & purification , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Oocytes/cytology , Oocytes/drug effects , Phosphorylation , Progesterone/pharmacology , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Xenopus
8.
FEBS Lett ; 490(1-2): 49-53, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11172809

ABSTRACT

Inwardly rectifying potassium channels require binding of phosphatidylinositol-4,5-bisphosphate (PIP2) for channel activity. Three independent sites (aa 175-206, aa 207-246, aa 324-365) were located in the C-terminal domain of Kir2.1 channels by assaying the binding of overlapping fragments to PIP2 containing liposomes. Mutations in the first site, which abolished channel activity, reduced PIP2 binding of this fragment but not of the complete C-terminus. Point mutations in the third site also reduced both, channel activity and PIP2 binding of this segment. The relevance of the third PIP2 binding site provides a basis for the understanding of constitutively active Kir2 channels.


Subject(s)
Binding Sites , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/chemistry , Saccharomyces cerevisiae Proteins , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Cattle , Chickens , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , Liposomes/metabolism , Mice , Mutagenesis, Site-Directed , Mutation , Oocytes/metabolism , Point Mutation , Potassium Channels/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Xenopus laevis
9.
Eur J Neurosci ; 12(8): 2823-32, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10971624

ABSTRACT

Scorpion alpha-toxins from Leiurus quinquestriatus hebraeus, LqhII and LqhIII, are similarly toxic to mice when administered by a subcutaneous route, but in mouse brain LqhII is 25-fold more toxic. Examination of the two toxins effects in central nervous system (CNS), peripheral preparations and expressed sodium channels revealed the basis for their differential toxicity. In rat brain synaptosomes, LqhII binds with high affinity, whereas LqhIII competes only at high concentration for LqhII-binding sites in a voltage-dependent manner. LqhII strongly inhibits sodium current inactivation of brain rBII subtype expressed in HEK293 cells, whereas LqhIII is weakly active at 2 microM, suggesting that LqhIII affects sodium channel subtypes other than rBII in the brain. In the periphery, both toxins inhibit tetrodotoxin-sensitive sodium current inactivation in dorsal root ganglion neurons, and are strongly active directly on the muscle and on expressed muI channels. Only LqhII, however, induced repetitive end-plate potentials in mouse phrenic nerve-hemidiaphragm muscle preparation by direct effect on the motor nerve. Thus, rBII and sodium channel subtypes expressed in peripheral nervous system (PNS) serve as the main targets for LqhII but are mostly not sensitive to LqhIII. Toxicity of both toxins in periphery may be attributed to the direct effect on muscle. Our data elucidate, for the first time, how different toxins affect mammalian central and peripheral excitable cells, and reveal unexpected subtype specificity of toxins that interact with receptor site 3.


Subject(s)
Brain Chemistry/physiology , Ion Channel Gating/drug effects , Phrenic Nerve/chemistry , Scorpion Venoms/pharmacology , Sodium Channels/metabolism , Animals , Binding Sites/physiology , Brain/cytology , Cells, Cultured , Female , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Humans , Ion Channel Gating/physiology , Kidney/cytology , Mammals , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Molecular Sequence Data , Motor Neurons/chemistry , Motor Neurons/cytology , Motor Neurons/drug effects , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , Neuromuscular Junction/chemistry , Neuromuscular Junction/cytology , Neurons, Afferent/chemistry , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Patch-Clamp Techniques , Phrenic Nerve/cytology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Scorpion Venoms/metabolism , Sequence Homology, Amino Acid , Sodium Channels/chemistry , Synaptosomes/chemistry , Synaptosomes/drug effects , Synaptosomes/physiology
10.
EMBO J ; 19(13): 3263-71, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10880439

ABSTRACT

Intracellular Ca(2+) inhibits voltage-gated potassium channels of the ether à go-go (EAG) family. To identify the underlying molecular mechanism, we expressed the human version hEAG1 in Xenopus oocytes. The channels lost Ca(2+) sensitivity when measured in cell-free membrane patches. However, Ca(2+) sensitivity could be restored by application of recombinant calmodulin (CaM). In the presence of CaM, half inhibition of hEAG1 channels was obtained in 100 nM Ca(2+). Overlay assays using labelled CaM and glutathione S-transferase (GST) fusion fragments of hEAG1 demonstrated direct binding of CaM to a C-terminal domain (hEAG1 amino acids 673-770). Point mutations within this section revealed a novel CaM-binding domain putatively forming an amphipathic helix with both sides being important for binding. The binding of CaM to hEAG1 is, in contrast to Ca(2+)-activated potassium channels, Ca(2+) dependent, with an apparent K(D) of 480 nM. Co-expression experiments of wild-type and mutant channels revealed that the binding of one CaM molecule per channel complex is sufficient for channel inhibition.


Subject(s)
Calcium/pharmacology , Calmodulin/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Trans-Activators , Amino Acid Sequence , Animals , Binding Sites , Calcium/metabolism , Calmodulin/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Ion Channel Gating , Molecular Sequence Data , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Binding , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transcriptional Regulator ERG , Xenopus laevis
11.
J Membr Biol ; 171(2): 107-15, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10489423

ABSTRACT

Ion channels and intracellular Ca2+ are thought to be involved in cell proliferation and may play a role in tumor development. We therefore characterized Ca(2+)-regulated potassium channels in the human melanoma cell lines IGR1, IPC298, and IGR39 using electrophysiological and molecular biological methods. All cell lines expressed outwardly rectifying K+ channels. Rapidly activating delayed rectifier channels were detected in IGR39 cells. The activation kinetics of voltage-gated K+ channels in IRG1 and IPC298 cells displayed characteristics of ether à go-go (eag) channels as they were much slower and depended both on the holding potential and on extracellular Mg2+. In addition, they could be blocked by physiological concentrations of intracellular Ca2+. In accordance with these electrophysiological results, analysis of mRNA revealed the expression of a gene coding for h-eag1 channels in IGR1 and IPC298 cells, but not in IGR39 cells. At elevated Ca2+ concentrations various types of Ca(2+)-activated K+ channels with single-channel characteristics similar to IK and SK channels were detected in IGR1 cells. The whole-cell Ca(2+)-activated K+ currents were not voltage dependent, insensitive for 100 nm apamin and 200 microm d-tubocurarine, but were blocked by charybdotoxin (100 nm) and clotrimazole (50 nm). Analysis of mRNA revealed the expression of hSK1, hSK2, and hIK channels in IGR1 cells.


Subject(s)
Calcium/metabolism , Melanoma/physiopathology , Potassium Channels/physiology , Humans , Ion Channel Gating , Patch-Clamp Techniques , Potassium Channels/analysis , RNA, Messenger/analysis , Tumor Cells, Cultured
12.
FEBS Lett ; 456(1): 17-21, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10452521

ABSTRACT

Oxidation of methionine residues in proteins to methionine sulfoxide can be reversed by the enzyme peptide methionine sulfoxide reductase (MsrA, EC 1.8.4.6). We cloned the gene encoding a human homologue (hMsrA) of the enzyme, which has an 88% amino acid sequence identity to the bovine version (bMsrA). With dot blot analyses based on RNA from human tissues, expression of hMsrA was found in all tissues tested, with highest mRNA levels in adult kidney and cerebellum, followed by liver, heart ventricles, bone marrow and hippocampus. In fetal tissue, expression was highest in the liver. No expression of hmsrA was detected in leukemia and lymphoma cell lines. To test if hMsrA is functional in cells, we assayed its effect on the inactivation time course of the A-type potassium channel ShC/B since this channel property strongly depends on the oxidative state of a methionine residue in the N-terminal part of the polypeptide. Co-expression of ShC/B and hMsrA in Xenopus oocytes significantly accelerated inactivation, showing that the cloned enzyme is functional in an in vivo assay system. Furthermore, the activity of a purified glutathione-S-transferase-hMsrA fusion protein was demonstrated in vitro by measuring the reduction of [3H]N-acetyl methionine sulfoxide.


Subject(s)
Oxidoreductases/genetics , Oxidoreductases/metabolism , Amino Acid Sequence , Animals , Cell Line/enzymology , Cerebellum/enzymology , Cloning, Molecular , Enzyme Activation , Female , Fetus/enzymology , Gene Expression Regulation, Developmental , Humans , Kidney/enzymology , Kidney/growth & development , Leukemia/enzymology , Liver/embryology , Liver/enzymology , Lung/enzymology , Lymphoma/enzymology , Methionine Sulfoxide Reductases , Molecular Sequence Data , Myocardium/enzymology , Oocytes/enzymology , Potassium Channels/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Xenopus laevis
13.
Pflugers Arch ; 438(2): 141-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10370099

ABSTRACT

Rat Kv1.4 potassium channels undergo rapid inactivation, which is mediated by the N-terminal structure of the polypeptide. This inactivation can be removed by N-terminal deletion of about 20 residues. However, more substantial deletion (e.g. 37 residues) restores inactivation suggesting a second inactivating domain [Kondoh et al. J Biol Chem 272:19333-19338, 1997]. Here we provide evidence that this inactivation shares all properties with N-type inactivation. Pore mutations, which are supposed to affect C-type inactivation, have no effect. In addition, the redox regulation of inactivation, which is typical for Kv1.4 channels, can be conferred to the inactivation of the deleted constructs by incorporation of an N-terminal cysteine residue. The most remarkable feature of this secondary inactivation is the existence of two components in the steady-state voltage dependence of inactivation. For mutant rKv1. 4Delta2-37 about 90% of the channels only activate when the holding membrane potential is more negative than about -120 mV; the remaining 10% show the typical threshold at -60 mV. Mutagenesis of the truncated channel affected the relative amplitudes of these two components, but not the voltage dependence. The results suggest that the secondary ball structures of rKv1.4 channels interact with the protein structures responsible for activation.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Animals , Electrophysiology , Kv1.4 Potassium Channel , Mutagenesis, Site-Directed , Potassium Channels/genetics , Sequence Deletion , Structure-Activity Relationship , Xenopus
14.
Eur J Neurosci ; 11(3): 753-60, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10103069

ABSTRACT

ERG (ether-à-go-go-related gene) K+ channels are crucial in human heart physiology (h-ERG), but are also found in neuronal cells and are impaired in Drosophila 'seizure' mutants. Their biophysical properties include the relatively fast kinetics of the inactivation gate and much slower kinetics of the activation gate. In order to elucidate how the complex time- and voltage-dependent activation properties of ERG channels underlies distinct roles in excitability, we investigated different types of ERG channels intrinsically present in cells or heterologously expressed in mammalian cells or Xenopus oocytes. Voltage-dependent activation curves were highly dependent on the features of the eliciting protocols. Only very long preconditioning times produced true steady-state relationships, a fact that has been largely neglected in the past, hampering the comparison of published data on ERG channels. Beyond this technical aspect, the slow activation property of ERG can be responsible for unsuspected physiological roles. We found that around the midpoint of the activation curve, the time constant of ERG open-close kinetics is of the order of 10-15 s. During sustained trains of depolarizations, e.g. those produced in neuronal firing, this leads to the use-dependent accumulation of open-state ERG channels. Accumulation is not observed in a mutant with a fast activation gate. In conclusion, it is well established that other K+ channels (i.e. Ca2+-activated and M) control the spike-frequency adaptation, but our results support the notion that the purely voltage-dependent activation property of ERG channels would allow a slow inhibitory physiological role in rapid neuronal signalling.


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Ion Channel Gating/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/metabolism , Trans-Activators , Animals , ERG1 Potassium Channel , Electric Stimulation , Electrophysiology , Ether-A-Go-Go Potassium Channels , Ganglia, Spinal/cytology , Humans , Kidney/cytology , Leukemia , Membrane Potentials/physiology , Mice , Mutagenesis/physiology , Neuroblastoma , Oocytes/physiology , Rats , Transcriptional Regulator ERG , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/physiology , Xenopus
15.
J Biol Chem ; 274(9): 5362-9, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10026145

ABSTRACT

The bovine ether à go-go gene encodes a delayed rectifier potassium channel. In contrast to other delayed rectifiers, its activation kinetics is largely determined by the holding potential and the concentration of extracellular Mg2+, giving rise to slowly activating currents with a characteristic sigmoidal rising phase. Replacement of a single amino acid in the extracellular linker between transmembrane segments S3 and S4 (L322H) strongly reduced the prepulse dependence and accelerated activation by 1 order of magnitude. In addition, compared with the wild type, the half-activation voltage of this mutant was shifted by more than 30 mV to more negative potentials. We used dimeric and tetrameric constructs of the bovine eag1 gene to analyze channels with defined stoichiometry of mutated and wild-type subunits within the tetrameric channel complexes. With increasing numbers of mutated subunits, the channel activation was progressively accelerated, and the sigmoidicity of the current traces was reduced. Based on a quantitative analysis, we show that the slow gating, typical for EAG channels, is mediated by independent conformational transitions of individual subunits, which gain their voltage dependence from the S4 segment. At a given voltage, external Mg2+ increases the probability of a channel subunit to be in the slowly activating conformation, whereas mutation L322H strongly reduces this probability.


Subject(s)
Ion Channel Gating , Potassium Channels/physiology , Animals , Cattle , Ether-A-Go-Go Potassium Channels , Kinetics , Magnesium/pharmacology , Membrane Potentials , Mutagenesis, Site-Directed , Potassium Channels/drug effects , Potassium Channels/genetics
16.
Nephrol Dial Transplant ; 13(9): 2234-40, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9761502

ABSTRACT

BACKGROUND: Renal functional prognosis is consistently more adverse in male individuals with renal disease. Male animals develop more marked proteinuria and glomerulosclerosis in several models of renal damage. Renal and glomerular growth are important permissive factors for progression of renal failure. PURPOSE OF THE STUDY: To investigate the influence of testosterone on renal and glomerular growth. DESIGN: Renal compensatory growth after uninephrectomy (UNX) was chosen as a model of renal growth. The effect of testosterone was assessed in control male, in orchidectomized (OX) male, and in ovariectomized (OV) female SD rats. Observation time was 10 months. MEASUREMENTS: Albuminuria by nephelometry; glomerular diameter, glomerular tuft area, renal zonal analysis by quantitative stereology. Testosterone and dihydroxytestosterone by gas chromatography and RIA. RESULTS: In sham-operated male rats, testosterone administration did not change the (left) kidney:body-weight ratio after uninephrectomy. In contrast, in OX male rats, testosterone administration caused a significant increase in kidney:body-weight ratio and in albuminuria. In these animals, glomerular diameter and outer stripe width were significantly lower in OX rats than in sham-operated controls. Glomerular volume and outer stripe width in OX animals were significantly higher after uninephrectomy (UNX) and were further increased in OX-UNX animals by administration of testosterone. Similar effects on glomerular diameter, cortical width (single) kidney:body-weight ratio were seen when OV female rats were treated with testosterone. CONCLUSION: After gonadal ablation, administration of testosterone amplifies compensatory glomerular and tubular growth in uninephrectomized male and female rats, i.e. testosterone is a permissive factor. Stimulation of glomerular growth may favour development of glomerulosclerosis.


Subject(s)
Kidney Glomerulus/drug effects , Nephrectomy/methods , Testosterone/pharmacology , Adaptation, Physiological/drug effects , Animals , Female , Kidney Glomerulus/growth & development , Male , Orchiectomy , Ovariectomy , Postoperative Period , Rats , Rats, Sprague-Dawley
17.
FEBS Lett ; 414(2): 435-8, 1997 Sep 08.
Article in English | MEDLINE | ID: mdl-9315735

ABSTRACT

The class III antiarrhythmic drug clofilium is known to block diverse delayed rectifier K+ channels at micromolar concentrations. In the present study we investigated the potency of clofilium and its tertiary analog LY97241 to inhibit K+ channels, encoded by the human ether-a-go-go related gene (HERG). Clofilium blocked HERG channels in a voltage-dependent fashion with an IC50 of 250 nM and 150 nM at 0 and +40 mV, respectively. LY97241 was almost 10-fold more potent (IC50 of 19 nM at +40 mV). Other cloned K+ channels which are also expressed in cardiac tissue, Kv1.1, Kv1.2, Kv1.4, Kv1.5, Kv4.2, Kir2.1, or I(Ks), were not affected by 100-fold higher concentrations. Block of HERG channels by LY97241 was voltage dependent and the rate of HERG inactivation was increased by LY97241. A rise of [K+]0 decreased both, rate of HERG inactivation and LY97241 affinity. The HERG S631A and S620T mutant channels which have a strongly reduced degree of inactivation were 7-fold and 33-fold less sensitive to LY97241 blockade, indicating that LY97241 binding is affected by HERG channel inactivation. In summary, the antiarrhythmic action of clofilium and its analog LY97241 appears to be caused by their potent, but distinct ability for blocking HERG channels.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Quaternary Ammonium Compounds/pharmacology , Trans-Activators , Animals , Cloning, Molecular , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Female , Humans , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutagenesis, Site-Directed , Oocytes/physiology , Point Mutation , Potassium Channels/biosynthesis , Potassium Channels/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Shal Potassium Channels , Structure-Activity Relationship , Transcriptional Regulator ERG , Xenopus laevis
18.
Br J Pharmacol ; 120(5): 968-74, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9138706

ABSTRACT

1. The antipsychotic drug haloperidol can induce a marked QT prolongation and polymorphic ventricular arrhythmias. In this study, we expressed several cloned cardiac K+ channels, including the human ether-a-go-go related gene (HERG) channels, in Xenopus oocytes and tested them for their haloperidol sensitivity. 2. Haloperidol had only little effects on the delayed rectifier channels Kv1.1, Kv1.2, Kv1.5 and IsK, the A-type channel Kv1.4 and the inward rectifier channel Kir2.1 (inhibition < 6% at 3 microM haloperidol). 3. In contrast, haloperidol blocked HERG channels potently with an IC50 value of approximately 1 microM. Reduced haloperidol, the primary metabolite of haloperidol, produced a block with an IC50 value of 2.6 microM. 4. Haloperidol block was use- and voltage-dependent, suggesting that it binds preferentially to either open or inactivated HERG channels. As haloperidol increased the degree and rate of HERG inactivation, binding to inactivated HERG channels is suggested. 5. The channel mutant HERG S631A has been shown to exhibit greatly reduced C-type inactivation which occurs only at potentials greater than 0 mV. Haloperidol block of HERG S631A at 0 mV was four fold weaker than for HERG wild-type channels. Haloperidol affinity for HERG S631A was increased four fold at +40 mV compared to 0 mV. 6. In summary, the data suggest that HERG channel blockade is involved in the arrhythmogenic side effects of haloperidol. The mechanism of haloperidol block involves binding to inactivated HERG channels.


Subject(s)
Antipsychotic Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Haloperidol/pharmacology , Long QT Syndrome/metabolism , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Trans-Activators , Animals , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Ion Channel Gating , Kinetics , Oocytes/metabolism , Potassium Channels/physiology , RNA, Complementary , Recombinant Proteins/antagonists & inhibitors , Transcriptional Regulator ERG , Xenopus
19.
J Physiol ; 493 ( Pt 3): 635-42, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8799887

ABSTRACT

1. The human eag-related potassium channel, HERG, gives rise to inwardly rectifying K+ currents when expressed in Xenopus oocytes. 2. The apparent inward rectification is caused by rapid inactivation. In extracellular Cs+ solutions, large outward currents can be recorded having an inactivation time constant at 0 mV of about 50 ms with an e-fold change every 37 mV. 3. HERG channel inactivation is not caused by an amino-terminal ball structure, as a deletion of the cytoplasmic amino terminus (HERG delta 2-373) did not eliminate inactivation. However, channel deactivation was accelerated about 12-fold at -80 mV. 4. Mutation of S631 to A, the homologous residue of eag channels, in the outer mouth of the HERG pore completely abolished channel inactivation. 5. Activity of HERG channels depended on extracellular cations, which are effective for channel activation, in the order Cs+ > K+ > > Li+ > Na+. The point mutation S631A strongly reduced this channel regulation. 6. By analogy to functional aspects of cloned voltage-gated potassium channels, rectification of HERG, as well as its kinetic properties during the course of an action potential, are presumably governed by a mechanism reminiscent of C-type inactivation.


Subject(s)
Potassium Channels/biosynthesis , Alanine/metabolism , Animals , Biotransformation/physiology , Humans , Ion Channel Gating/physiology , Kinetics , Membrane Potentials/physiology , Mutation/physiology , Oocytes/metabolism , Patch-Clamp Techniques , Polymerase Chain Reaction , Potassium Channels/genetics , Sequence Deletion/physiology , Serine/metabolism , Xenopus laevis
20.
Pflugers Arch ; 431(4): 483-93, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8596690

ABSTRACT

Shaker potassium channels undergo a slow C-type inactivation which can be hastened dramatically by single-point mutations in or near the pore region. We found that the oxidizing agent chloramine-T (Chl-T) causes an irreversible loss of current for those mutants which show C-type inactivation. For several mutants at position T449, which show a wide spectrum of inactivation time constants, the time constant of current rundown induced by Chl-T correlated with the speed of inactivation. Rundown was accelerated when the channels were in the inactivated state but rundown also occurred when channels were not opened or inactivated. Apparently, only those channels which can undergo C-type inactivation are accessible to Chl-T. In order to gain information about the target amino-acid residue for the action of Chl-T and the structural rearrangements occurring during C-type inactivation, several mutant channel proteins were compared with respect to their response to Chl-T. Since Chl-T can oxidize cysteine and methionine residues, we mutated the possible targets in and close to the pore region, namely C462 to A, and M440 and M448 to I. While the residues M440 and C462 were not important for channel rundown, mutation of M448 to I made the channels more resistant to Chl-T by about one order of magnitude. While inactivation was accelerated upon application of Chl-T in most mutants, mutation of M448 to I abolished this effect on the time course of inactivation, indicating that M448 is one of the target residues for Chl-T.


Subject(s)
Chloramines/pharmacology , Potassium Channels/physiology , Tosyl Compounds/pharmacology , Animals , Anti-Infective Agents/pharmacology , Dose-Response Relationship, Drug , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/physiology , Mutation/drug effects , Potassium Channels/genetics , Time Factors , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...