PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 27176799-0 2016 Macrolide antibiotics exert antileukemic effects by modulating the autophagic flux through inhibition of hERG1 potassium channels. macrolide antibiotics 0-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-110 26548443-2 2016 We describe two cases where a switch to bisoprolol resulted in worsening of arrhythmia control: A man with LQT2, asymptomatic on propranolol, experienced syncope after switching to bisoprolol 5 mg daily. Bisoprolol 40-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-111 26548443-2 2016 We describe two cases where a switch to bisoprolol resulted in worsening of arrhythmia control: A man with LQT2, asymptomatic on propranolol, experienced syncope after switching to bisoprolol 5 mg daily. Propranolol 129-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-111 26271031-0 2016 Estradiol regulates human QT-interval: acceleration of cardiac repolarization by enhanced KCNH2 membrane trafficking. Estradiol 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-95 26271031-12 2016 Estradiol enhanced the physical interaction of KCNH2-channel subunits with heat-shock proteins and augmented ion-channel trafficking to the membrane. Estradiol 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-52 26271031-14 2016 Estradiol acts on KCNH2 channels via enhanced estradiol-receptor-alpha-mediated Hsp90 interaction, augments membrane trafficking and thereby increases repolarizing current. Estradiol 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-23 26481172-0 2016 Differential Response to Risperidone in Schizophrenia Patients by KCNH2 Genotype and Drug Metabolizer Status. Risperidone 25-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-71 27013874-2 2016 In light of this goal, we report a robust one-step method for the synthesis of dicarboxylic acid-terminated polyethylene glycol (PEG)-gold nanoparticles (AuNPs) and doxorubicin-loaded PEG-AuNPs, and their further antibody targeting (anti-Kv11.1 polyclonal antibody [pAb]). Dicarboxylic Acids 79-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 238-244 27013874-2 2016 In light of this goal, we report a robust one-step method for the synthesis of dicarboxylic acid-terminated polyethylene glycol (PEG)-gold nanoparticles (AuNPs) and doxorubicin-loaded PEG-AuNPs, and their further antibody targeting (anti-Kv11.1 polyclonal antibody [pAb]). Polyethylene Glycols 129-132 potassium voltage-gated channel subfamily H member 2 Homo sapiens 238-244 27013874-2 2016 In light of this goal, we report a robust one-step method for the synthesis of dicarboxylic acid-terminated polyethylene glycol (PEG)-gold nanoparticles (AuNPs) and doxorubicin-loaded PEG-AuNPs, and their further antibody targeting (anti-Kv11.1 polyclonal antibody [pAb]). Doxorubicin 165-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 238-244 26481172-7 2016 RESULTS: Risperidone caused greater in vitro block of the alternatively spliced Kv11.1-3.1 isoform than full-length Kv11.1-1A channels, whereas its metabolite paliperidone and other atypical antipsychotics have similar potencies for the two isoforms. Risperidone 9-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-86 26481172-8 2016 In the CATIE study (N=362), patients with genotypes associated with increased Kv11.1-3.1 expression (N=52) showed a better treatment response to risperidone compared with other drugs, but this association was dependent on metabolism status. Risperidone 145-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-84 26481172-9 2016 Patients with KCNH2 risk genotypes and slow metabolizer status (approximately 7% of patients) showed marked improvement in symptoms when treated with risperidone compared with patients with fast metabolizer status or without the KCNH2 risk genotypes. Risperidone 150-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-19 26481172-10 2016 CONCLUSIONS: These data support the hypothesis that Kv11.1 channels play a role in the therapeutic action of antipsychotic drugs, particularly risperidone, and further highlight the promise of optimizing response with genotype-guided therapy for schizophrenia patients. Risperidone 143-154 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-58 26068526-6 2016 Electrophysiological studies showed that macrolides prolonging the QT interval inhibit the rapid component of the delayed rectifier K(+) current (IKr) through the block of potassium channels encoded by the human ether-a-go-go-related gene (HERG). Macrolides 41-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 240-244 27069917-9 2016 By facilitating expression of channel protein HERG, potassium ions may prevent it from being shunted to procancerous pathways by inducing apoptosis. Potassium 52-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 27069917-11 2016 Thus, our findings suggest that potassium ions could inhibit tumorigenesis through inducing apoptosis of hepatoma cells by upregulating potassium ions transport channel proteins HERG and VDAC1. Potassium 32-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 26649323-0 2016 HERG Protein Plays a Role in Moxifloxacin-Induced Hypoglycemia. Moxifloxacin 29-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 26649323-1 2016 The purpose of this study was to investigate the effect of moxifloxacin on HERG channel protein and glucose metabolism. Moxifloxacin 59-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-79 26649323-3 2016 The whole-cell patch clamp method was used to examine the effect of moxifloxacin on HERG channel currents. Moxifloxacin 68-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 26649323-6 2016 Moxifloxacin inhibited HERG time-dependent and tail currents in HEK293 cells in a concentration-dependent manner. Moxifloxacin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 26649323-10 2016 Serum glucose levels increased and insulin concentrations decreased in HERG knockout mice when compared to wild-type mice. Glucose 6-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 26649323-11 2016 The moxifloxacin-induced decrease in blood glucose and increase in insulin secretion occurred via the HERG protein; thus, HERG protein plays a role in insulin secretion. Moxifloxacin 4-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 26649323-11 2016 The moxifloxacin-induced decrease in blood glucose and increase in insulin secretion occurred via the HERG protein; thus, HERG protein plays a role in insulin secretion. Moxifloxacin 4-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 26277508-2 2015 METHODS: HEK293 cells were transiently transfected with HERG channel cDNA plasmid pcDNA3.1 via Lipofectamine. Lipofectamine 95-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 26503718-6 2015 Open-state stabilization by PIP2 has been observed in human Erg1, but the proposed site of regulation in the distal C terminus is not conserved among EAG family channels. Phosphatidylinositol 4,5-Diphosphate 28-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 26376488-11 2015 Furthermore, the equine KV11.1 channel was susceptible to pharmacological block with terfenadine. Terfenadine 85-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-30 25790957-6 2015 Experiments using the KCNH2-pore blocking agent quinidine supported these findings. Quinidine 48-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-27 25986146-0 2015 Ivabradine prolongs phase 3 of cardiac repolarization and blocks the hERG1 (KCNH2) current over a concentration-range overlapping with that required to block HCN4. Ivabradine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-74 25986146-0 2015 Ivabradine prolongs phase 3 of cardiac repolarization and blocks the hERG1 (KCNH2) current over a concentration-range overlapping with that required to block HCN4. Ivabradine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-81 25986146-5 2015 The objective of this study is to assess whether ivabradine blocked the hERG1 current. Ivabradine 49-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 25986146-9 2015 Disruption of C-type inactivation also suppressed block of hERG1 by ivabradine. Ivabradine 68-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-64 25986146-10 2015 Molecular docking and molecular dynamics simulations indicate that ivabradine may access the inner cavity of the hERG1 via a lipophilic route and has a well-defined binding site in the closed state of the channel. Ivabradine 67-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-118 25888115-6 2015 Analysis of a panel of mutant hERG1 and rERG2 channels further revealed that seven residues, five in the C-linker and two in the adjacent region of the cyclic nucleotide-binding homology domain, can fully account for the differential sensitivity of hERG1 and rERG2 channels to RPR. Nucleotides, Cyclic 152-169 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-35 25888115-6 2015 Analysis of a panel of mutant hERG1 and rERG2 channels further revealed that seven residues, five in the C-linker and two in the adjacent region of the cyclic nucleotide-binding homology domain, can fully account for the differential sensitivity of hERG1 and rERG2 channels to RPR. Nucleotides, Cyclic 152-169 potassium voltage-gated channel subfamily H member 2 Homo sapiens 249-254 26076856-0 2015 QT interval prolongation in a patient with LQT2 on levetiracetam. Levetiracetam 51-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 25855787-4 2015 Concatenated hERG1 tetramers containing four wild-type subunits exhibited high-affinity block by cisapride, dofetilide, and MK-499, similar to wild-type channels formed from hERG1 monomers. Cisapride 97-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-18 25855787-4 2015 Concatenated hERG1 tetramers containing four wild-type subunits exhibited high-affinity block by cisapride, dofetilide, and MK-499, similar to wild-type channels formed from hERG1 monomers. dofetilide 108-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-18 25855787-4 2015 Concatenated hERG1 tetramers containing four wild-type subunits exhibited high-affinity block by cisapride, dofetilide, and MK-499, similar to wild-type channels formed from hERG1 monomers. L 706000 124-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-18 25800797-6 2015 HEK293 cells stably transfected with KCNH2 (hERG) cDNA were used to examine the effects of hypaconitine on the KCNH2 channel by using the manual patch clamp technique. hypaconitine 91-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 111-116 25800797-10 2015 In the in vitro study in HEK293 cells, hypaconitine inhibited the KCNH2 currents in a concentration-dependent manner with an IC50 of 8.1nM. hypaconitine 39-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-71 25911606-2 2015 This study investigated the propensity of ivabradine to interact with KCNH2-encoded human Ether-a-go-go-Related Gene (hERG) potassium channels, which strongly influence ventricular repolarization and susceptibility to torsades de pointes arrhythmia. Ivabradine 42-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-75 25967940-9 2015 The best combination of in silico tools for KCNH2 is SIFT and PROVEAN or PROVEAN, SNPs&GO and SIFT. Adenosine Monophosphate 87-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-49 26937396-3 2015 Changes were identified in the KCNH2 gene and mitochondrial tRNA for cysteine. Cysteine 69-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-36 25719829-6 2015 ERG1 expression in vivo was determined by optical imaging using Alexa-680-labelled alpha-hERG1-MoAb. alexa-680 64-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-94 25468537-4 2015 Norsandwicine, 10-methoxypanarine, tueiaoine, and more importantly nukuhivensiums, were shown to significantly induce a reduction of IKr amplitude (HERG current). norsandwicine 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 25576780-11 2015 The second AED rescue occurred in a remotely symptomatic 14-year-old boy with high-risk LQT2 (QTc >550 ms) on a beta-blocker who previously declined a prophylactic ICD. qtc 94-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 25296617-4 2015 EXPERIMENTAL APPROACH: The affinity and kinetic parameters of 15 prototypical Kv 11.1 inhibitors were evaluated in a number of [(3) H]-dofetilide binding assays. [(3) h]-dofetilide 127-145 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-85 25296617-6 2015 KEY RESULTS: A novel [(3) H]-dofetilide competition association assay was set up and validated, which allowed us to determine the binding kinetics of the Kv 11.1 blockers used in this study. [(3) h]-dofetilide 21-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 154-161 25596745-0 2015 Stimulation of hERG1 channel activity promotes a calcium-dependent degradation of cyclin E2, but not cyclin E1, in breast cancer cells. Calcium 49-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-20 25596745-4 2015 In addition we have also reveal that hERG1 stimulation induces an increase in intracellular calcium that is required for cyclin E2 degradation. Calcium 92-99 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-42 25704028-0 2015 Phosphatidylinositol4-phosphate 5-kinase prevents the decrease in the HERG potassium current induced by Gq protein-coupled receptor stimulation. Potassium 75-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 25704028-1 2015 The human ether-a-go-go-related gene (HERG) potassium current (IHERG) has been shown to decrease in amplitude following stimulation with Gq protein-coupled receptors (GqRs), such as alpha1-adrenergic and M1-muscarinic receptors (alpha1R and M1R, respectively), at least partly via the reduction of membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). Potassium 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 25704028-1 2015 The human ether-a-go-go-related gene (HERG) potassium current (IHERG) has been shown to decrease in amplitude following stimulation with Gq protein-coupled receptors (GqRs), such as alpha1-adrenergic and M1-muscarinic receptors (alpha1R and M1R, respectively), at least partly via the reduction of membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). Phosphatidylinositol 4,5-Diphosphate 307-344 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 25704028-1 2015 The human ether-a-go-go-related gene (HERG) potassium current (IHERG) has been shown to decrease in amplitude following stimulation with Gq protein-coupled receptors (GqRs), such as alpha1-adrenergic and M1-muscarinic receptors (alpha1R and M1R, respectively), at least partly via the reduction of membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). pi(4,5)p2 346-355 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 25704028-2 2015 The present study was designed to investigate the modulation of HERG channels by PI(4,5)P2 and phosphatidylinositol4-phosphate 5-kinase (PI(4)P5-K), a synthetic enzyme of PI(4,5)P2. pi(4,5)p2 81-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 25704028-2 2015 The present study was designed to investigate the modulation of HERG channels by PI(4,5)P2 and phosphatidylinositol4-phosphate 5-kinase (PI(4)P5-K), a synthetic enzyme of PI(4,5)P2. pi(4,5)p2 171-180 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 25468537-4 2015 Norsandwicine, 10-methoxypanarine, tueiaoine, and more importantly nukuhivensiums, were shown to significantly induce a reduction of IKr amplitude (HERG current). 10-methoxypanarine 15-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 25468537-4 2015 Norsandwicine, 10-methoxypanarine, tueiaoine, and more importantly nukuhivensiums, were shown to significantly induce a reduction of IKr amplitude (HERG current). tueiaoine 35-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 25232191-11 2014 Considering the substantial relationship between LQT2 and epilepsy, these findings reveal that NS1643 is a useful compound to elucidate the causal connection of LQT2 and epilepsy. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 95-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 25232191-11 2014 Considering the substantial relationship between LQT2 and epilepsy, these findings reveal that NS1643 is a useful compound to elucidate the causal connection of LQT2 and epilepsy. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 95-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 161-165 25974115-4 2015 A novel mutation KCNH2-I560T, when expressed in COS-7 cells, showed a 2.5-fold increase in peak current density, and a positive shift (+14 mV) of the inactivation curve compared with wild type. carbonyl sulfide 48-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-22 25100024-0 2014 Molecular docking and molecular dynamics studies on the structure-activity relationship of fluoroquinolone for the HERG channel. Fluoroquinolones 91-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 25249569-8 2014 Stimulation of LQT2 cells with beta-adrenergic agonist isoproterenol resulted in prolongation of the plateau of action potentials accompanied by aberrant Ca(2+) releases and EADs, which were abolished by inhibition of Ca(2+)/calmodulin-dependent protein kinase type 2. Isoproterenol 55-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 25100024-1 2014 Fluoroquinolones play an important role in the treatment of serious bacterial infections, but at the same time they could lead to cardiac toxicity due to the blockage of the HERG potassium channel, which even leads to the withdrawal of some fluoroquinolones. Fluoroquinolones 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 25100024-1 2014 Fluoroquinolones play an important role in the treatment of serious bacterial infections, but at the same time they could lead to cardiac toxicity due to the blockage of the HERG potassium channel, which even leads to the withdrawal of some fluoroquinolones. Fluoroquinolones 241-257 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 25128783-0 2014 Upregulation of functional Kv11.1 isoform expression by inhibition of intronic polyadenylation with antisense morpholino oligonucleotides. Morpholinos 110-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-33 25128783-6 2014 More importantly, blocking these elements by antisense morpholino oligonucleotides shifted the alternative processing of KCNH2 intron 9 from the polyadenylation to the splicing pathway, leading to the predominant production of Kv11.1a and a significant increase in Kv11.1 current. Morpholinos 55-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-126 25128783-6 2014 More importantly, blocking these elements by antisense morpholino oligonucleotides shifted the alternative processing of KCNH2 intron 9 from the polyadenylation to the splicing pathway, leading to the predominant production of Kv11.1a and a significant increase in Kv11.1 current. Morpholinos 55-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 227-233 25257637-7 2014 In LQT1, the risk reduction for first cardiac events was similar among the 4 beta-blockers, but in LQT2, nadolol provided the only significant risk reduction (hazard ratio: 0.40 [0.16 to 0.98]). Nadolol 105-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 25257637-9 2014 CONCLUSIONS: Although the 4 beta-blockers are equally effective in reducing the risk of a first cardiac event in LQTS, their efficacy differed by genotype; nadolol was the only beta-blocker associated with a significant risk reduction in patients with LQT2. Nadolol 156-163 potassium voltage-gated channel subfamily H member 2 Homo sapiens 252-256 25100024-3 2014 Though there were large amounts of bioactivity data of fluoroquinolones on the blockage of HERG, little structural basis of binding of blockers to the HERG channel was known. Fluoroquinolones 55-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 25100024-4 2014 Here, we combined molecular docking, molecular dynamics simulations, free energy calculations and binding energy decomposition analysis to explore the binding modes of fluoroquinolones in the HERG potassium channel. Fluoroquinolones 168-184 potassium voltage-gated channel subfamily H member 2 Homo sapiens 192-196 25100024-6 2014 Our results showed that the CH3 group in MX was favorable for the binding to the HERG channel, while Tyr652 and Phe656 were critical for the hydrophobic interaction between fluoroquinolones and the HERG channel. Fluoroquinolones 173-189 potassium voltage-gated channel subfamily H member 2 Homo sapiens 198-202 24846011-0 2014 Inhibition of HERG potassium channels by domiphen bromide and didecyl dimethylammonium bromide. domiphen 41-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 24846011-0 2014 Inhibition of HERG potassium channels by domiphen bromide and didecyl dimethylammonium bromide. didecyldimethylammonium 62-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 24846011-1 2014 Domiphen bromide and didecyl dimethylammonium bromide were widely used environmental chemicals with potent activity on blockade of human ether-a-go-go related gene (HERG) channels. domiphen 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-169 24846011-1 2014 Domiphen bromide and didecyl dimethylammonium bromide were widely used environmental chemicals with potent activity on blockade of human ether-a-go-go related gene (HERG) channels. didecyldimethylammonium 21-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-169 24846011-3 2014 The kinetics of block of HERG channels by domiphen bromide and didecyl dimethylammonium bromide was studied in order to characterize the inhibition of HERG currents by these quaternary ammonium compounds (QACs). domiphen 42-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 24846011-3 2014 The kinetics of block of HERG channels by domiphen bromide and didecyl dimethylammonium bromide was studied in order to characterize the inhibition of HERG currents by these quaternary ammonium compounds (QACs). didecyldimethylammonium 63-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 24846011-3 2014 The kinetics of block of HERG channels by domiphen bromide and didecyl dimethylammonium bromide was studied in order to characterize the inhibition of HERG currents by these quaternary ammonium compounds (QACs). Quaternary Ammonium Compounds 174-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 24846011-3 2014 The kinetics of block of HERG channels by domiphen bromide and didecyl dimethylammonium bromide was studied in order to characterize the inhibition of HERG currents by these quaternary ammonium compounds (QACs). Quaternary Ammonium Compounds 174-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-155 24846011-4 2014 Domiphen bromide and didecyl dimethylammonium bromide inhibited HERG channel currents in a dose-dependent manner with IC50 values of 9nM and 5nM, respectively. domiphen 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 24846011-4 2014 Domiphen bromide and didecyl dimethylammonium bromide inhibited HERG channel currents in a dose-dependent manner with IC50 values of 9nM and 5nM, respectively. didecyldimethylammonium 21-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 24846011-5 2014 Block of HERG channel by domiphen bromide and didecyl dimethylammonium bromide was voltage-dependent and use-dependent. domiphen 25-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 24846011-5 2014 Block of HERG channel by domiphen bromide and didecyl dimethylammonium bromide was voltage-dependent and use-dependent. didecyldimethylammonium 46-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 24846011-7 2014 The docking models implied that these two compounds bound to PAS domain of HERG channels and inhibited its function. Protactinium 61-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-79 24846011-8 2014 Our data demonstrated that domiphen bromide and didecyl dimethylammonium bromide blocked the HERG channel with a preference for the activated channel state. domiphen 27-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 24846011-8 2014 Our data demonstrated that domiphen bromide and didecyl dimethylammonium bromide blocked the HERG channel with a preference for the activated channel state. didecyldimethylammonium 48-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 24464434-0 2014 Roles of PKC Isoforms in PMA-Induced Modulation of the hERG Channel (Kv11.1). Tetradecanoylphorbol Acetate 25-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-75 25254341-4 2014 In myocytes carrying an LQT2 mutation (HERG-A422T), APs and [Ca(2+)]i transients were prolonged in parallel. Adenosine Phosphosulfate 52-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 25254341-4 2014 In myocytes carrying an LQT2 mutation (HERG-A422T), APs and [Ca(2+)]i transients were prolonged in parallel. Adenosine Phosphosulfate 52-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 24875677-4 2014 In order to evaluate if any polymorphism of potassium channels" genes could explain some of the "idiosyncratic" QT prolongations observed in patients treated with methadone, we tested the association between KCNE1, KCNE2, and KCNH2 polymorphism and the QT interval prolongation in those patients, controlling for other variables associated with a decrease of the repolarizing reserve. Methadone 163-172 potassium voltage-gated channel subfamily H member 2 Homo sapiens 226-231 24875677-9 2014 CONCLUSION: KCNH2 genotyping may be relevant in the analysis of cumulative risk factors for QT prolongation in patients on methadone maintenance treatment. Methadone 123-132 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-17 24638994-4 2014 In human ether-a-go-go-related gene 1 (hERG1) K(+) channels, C-type inactivation is allosterically inhibited by ICA-105574, a substituted benzamide. isocyanic acid 112-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-44 24630832-2 2014 In experimental systems varying from Drosophila to primary mammalian and human cell lines, celecoxib inhibits many voltage-activated Na(+), Ca(2+), and K(+) channels, including NaV1.5, L- and T-type Ca(2+) channels, KV1.5, KV2.1, KV4.3, KV7.1, KV11.1 (hERG), while stimulating other K(+) channels-KV7.2-5 and, possibly, KV11.1 (hERG) channels under certain conditions. Celecoxib 91-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 244-250 24630832-2 2014 In experimental systems varying from Drosophila to primary mammalian and human cell lines, celecoxib inhibits many voltage-activated Na(+), Ca(2+), and K(+) channels, including NaV1.5, L- and T-type Ca(2+) channels, KV1.5, KV2.1, KV4.3, KV7.1, KV11.1 (hERG), while stimulating other K(+) channels-KV7.2-5 and, possibly, KV11.1 (hERG) channels under certain conditions. Celecoxib 91-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 320-326 24530480-8 2014 Finally, we used a full-length hERG splicing-competent construct to show that inhibition of downstream intron splicing by antisense morpholino oligonucleotides inhibited NMD and rescued the functional expression of a third LQT2 mutation, Y1078. Morpholinos 132-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 223-227 24638994-4 2014 In human ether-a-go-go-related gene 1 (hERG1) K(+) channels, C-type inactivation is allosterically inhibited by ICA-105574, a substituted benzamide. benzamide 138-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-44 24638994-10 2014 Enhancement of hERG1 channel current magnitude by PD-118057 and attenuated inactivation by ICA-105574 were mediated by cooperative subunit interactions. 2-(4-(2-(3,4-dichlorophenyl)ethyl)phenylamino)benzoic acid 50-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-20 24638994-10 2014 Enhancement of hERG1 channel current magnitude by PD-118057 and attenuated inactivation by ICA-105574 were mediated by cooperative subunit interactions. isocyanic acid 91-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-20 24516604-4 2014 The small molecule hERG ligand doxazosin induced concentration-dependent apoptosis of human LNT-229 (EC50 = 35 microM) and U87MG (EC50 = 29 microM) GB cells, accompanied by cell cycle arrest in the G0/G1 phase. Doxazosin 31-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 24606761-1 2014 BACKGROUND: The human ether-a-go-go related gene 1 (hERG1), which codes for a potassium ion channel, is a key element in the cardiac delayed rectified potassium current, IKr, and plays an important role in the normal repolarization of the heart"s action potential. Potassium 78-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-57 24606761-4 2014 METHODS: In this proof-of-principle study, we focus on cisapride, a gastroprokinetic agent withdrawn from the market due to its high hERG1 blocking affinity. Cisapride 55-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-138 24606930-0 2014 Proline scan of the HERG channel S6 helix reveals the location of the intracellular pore gate. Proline 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 24366185-0 2014 Comparison of read-through effects of aminoglycosides and PTC124 on rescuing nonsense mutations of HERG gene associated with long QT syndrome. Aminoglycosides 38-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 24366185-2 2014 The read-through effects of different aminoglycosides and PTC124 on HERG gene have yet to be adequately elucidated. Aminoglycosides 38-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 24366185-13 2014 The above results suggest that aminoglycosides and PTC124 induced different effects on rescue nonsense mutations of the HERG gene. Aminoglycosides 31-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 24366185-13 2014 The above results suggest that aminoglycosides and PTC124 induced different effects on rescue nonsense mutations of the HERG gene. ataluren 51-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 24516604-6 2014 HERG suppression via siRNA-mediated knock down mimicked pro-apoptotic effects of doxazosin. Doxazosin 81-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 24212718-3 2014 Reduced serum glucose blocks the repolarizing K(+) channel HERG, which leads to action potential and QT prolongation and is uniformly associated with risk for torsades de pointes ventricular tachycardia. Glucose 14-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 24475014-4 2014 We employed AP-clamp techniques using physiological action potential waveforms recorded from various regions of canine heart to study HERG function in HEK293 cells and identified several novel aspects of HERG function. CLAmP 15-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 204-208 24475014-10 2014 Based on our results, we conclude that the distinct biophysical properties of HERG reported by AP-clamp confer its unique function in cardiac repolarization thereby in antiarrhythmia and arrhythmogenesis. ap-clamp 95-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 25177033-0 2014 Blockade of HERG human K+ channels by the antidepressant drug paroxetine. Paroxetine 62-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 24606761-7 2014 An analysis of the fragment interactions of cisapride at human A2A adenosine receptors and hERG1 central cavities helped us to identify the key chemical groups responsible for the drug activity and hERG1 blockade. Cisapride 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-96 24606761-7 2014 An analysis of the fragment interactions of cisapride at human A2A adenosine receptors and hERG1 central cavities helped us to identify the key chemical groups responsible for the drug activity and hERG1 blockade. Cisapride 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 198-203 24606761-10 2014 CONCLUSIONS: An interaction decomposition of cisapride and cisapride derivatives allowed for the identification of key active scaffolds and functional groups that may be responsible for the unwanted blockade of hERG1. Cisapride 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 211-216 24606761-10 2014 CONCLUSIONS: An interaction decomposition of cisapride and cisapride derivatives allowed for the identification of key active scaffolds and functional groups that may be responsible for the unwanted blockade of hERG1. Cisapride 59-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 211-216 25366487-0 2014 Effect of terfenadine and pentamidine on the HERG channel and its intracellular trafficking: combined analysis with automated voltage clamp and confocal microscopy. Terfenadine 10-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 25366487-0 2014 Effect of terfenadine and pentamidine on the HERG channel and its intracellular trafficking: combined analysis with automated voltage clamp and confocal microscopy. Pentamidine 26-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 25177033-1 2014 The effects of paroxetine, a selective serotonin reuptake inhibitor, on human ether-a-go-go-related gene (HERG) channels were investigated using the whole-cell patch-clamp technique. Paroxetine 15-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 25177033-3 2014 Paroxetine inhibited the peak tail currents of the HERG channel in a concentration-dependent manner, with an IC50 value of 0.45 microM and a Hill coefficient of 0.85. Paroxetine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 24386440-2 2013 METHODS: We treated neonatal rat ventricular myocytes and HEK293 cells with stable expression of h-ERG with H2O2 for 12 h and 48 h. Expression of miR-17-5p seed miRNAs was quantified by real-time RT-PCR. Hydrogen Peroxide 108-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-102 24386440-6 2013 RESULTS: H-ERG trafficking was impaired by H2O2 after 48 h treatment, accompanied by reciprocal changes of expression between miR-17-5p seed miRNAs and several chaperones (Hsp70, Hsc70, CANX, and Golga2), with the former upregulated and the latter downregulated. Hydrogen Peroxide 43-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-14 24386440-8 2013 Application miR-17-5p inhibitor rescued H2O2-induced impairment of h-ERG trafficking. Hydrogen Peroxide 40-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-72 24386440-9 2013 Upregulation of endogenous by H2O2 or forced miR-17-5p expression either reduced h-ERG current. Hydrogen Peroxide 30-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-86 24369231-2 2013 METHODS: The plasmids pcDNA3.1-PTPN12-RFP and herg mutant constructed by PCR technique were transfected into HEK293 cells via Lipofectamine 2000, and the cells stably expressing PTPN12 selected with G418 were identified by Western blotting with anti-PTPN12 antibody. Lipofectamine 126-144 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 25246284-6 2014 COX-2 inhibitor and PGE2 had influence on the HERG current in gastric cancer cells. Dinoprostone 20-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 25246284-7 2014 COX-2 inhibitor reduced the amplitude of HERG current in gastric cancer cells and PGE2 enhanced the amplitude. Dinoprostone 82-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 25246284-8 2014 However, in gastric cancer cells transfected with HERG mutant deleting cAMP-binding domain, both COX-2 inhibitor and PGE2 did not show significant effects on HERG current. Cyclic AMP 71-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 25246284-8 2014 However, in gastric cancer cells transfected with HERG mutant deleting cAMP-binding domain, both COX-2 inhibitor and PGE2 did not show significant effects on HERG current. Dinoprostone 117-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 25246284-9 2014 cAMP agonist enhanced the amplitude of HERG current and cAMP antagonist reduced the amplitude in gastric cancer cells. Cyclic AMP 0-4 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 25177033-5 2014 The paroxetine-induced inhibition of the HERG channels was voltage-dependent. Paroxetine 4-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 25177033-9 2014 Paroxetine induced a leftward shift in the voltage-dependence of the steady-state activation of the HERG channels. Paroxetine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 25177033-14 2014 These results suggest that paroxetine blocks the HERG channels by binding to these channels in the open and inactivated states. Paroxetine 27-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 24193004-6 2013 We here propose a method for hERG1 gene quantification in colorectal cancer samples in both cryopreserved and formalin-fixed and paraffin-embedded samples. Formaldehyde 110-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-34 24193004-6 2013 We here propose a method for hERG1 gene quantification in colorectal cancer samples in both cryopreserved and formalin-fixed and paraffin-embedded samples. Paraffin 129-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-34 24369231-7 2013 CONCLUSION: PTPN12 negatively regulates cardiac HERG channel cerrent possibly by decreasing the phosphorylation level of HERG tyrosine residues. Tyrosine 126-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 24369231-7 2013 CONCLUSION: PTPN12 negatively regulates cardiac HERG channel cerrent possibly by decreasing the phosphorylation level of HERG tyrosine residues. Tyrosine 126-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 24206565-8 2013 Potassium supplementation and sex hormone-based therapy may protect patients with LQT2. Potassium 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 23718892-0 2013 Differential effects of the beta-adrenoceptor blockers carvedilol and metoprolol on SQT1- and SQT2-mutant channels. Carvedilol 55-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 23864605-11 2013 These data suggest that pharmacological correction quickly increases the trafficking of LQT2 channels stored in the transitional ER via a Rab11B-dependent pathway, and we conclude that the pharmacological chaperone activity of drugs like ranolazine might have therapeutic potential. Ranolazine 238-248 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 23718892-0 2013 Differential effects of the beta-adrenoceptor blockers carvedilol and metoprolol on SQT1- and SQT2-mutant channels. Metoprolol 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 23718892-7 2013 Carvedilol"s (1 muM) inhibition of the IKr tail was attenuated in N588K-KCNH2 (4.5 +- 3% vs 50.3 +- 4%, WT, P < 0.001) with IC50 values of 2.8 muM (WT) and 25.4 muM (N588K). Carvedilol 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 23718892-8 2013 Carvedilol"s IKr end-pulse inhibition, however, was increased in N588K-KCNH2 (10 muM, 60.7 +- 6% vs 36.5 +- 5%, WT, P < 0.01). Carvedilol 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-76 23718892-10 2013 CONCLUSIONS: N588K-KCNH2 and V307L-KCNQ1 mutations decrease carvedilol"s inhibition of the IKs or IKr tail but increase carvedilol"s IKr end-pulse inhibition and metoprolol"s inhibition of tail and end-pulse currents. Carvedilol 60-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-24 23721480-2 2013 Previous studies have identified the cytosolic PAS (Per/Arnt/Sim) domain as a hotspot for mutations that cause Kv11.1 trafficking defects. Aminosalicylic Acid 47-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 111-117 23793622-0 2013 Cholesterol regulates HERG K+ channel activation by increasing phospholipase C beta1 expression. Cholesterol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 23793622-3 2013 Recently, we showed that enrichment of cell membrane with cholesterol inhibits HERG channels by reducing the levels of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] due to the activation of phospholipase C (PLC). Cholesterol 58-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 23793622-3 2013 Recently, we showed that enrichment of cell membrane with cholesterol inhibits HERG channels by reducing the levels of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] due to the activation of phospholipase C (PLC). Phosphatidylinositol 4,5-Diphosphate 119-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 23793622-3 2013 Recently, we showed that enrichment of cell membrane with cholesterol inhibits HERG channels by reducing the levels of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] due to the activation of phospholipase C (PLC). Phosphatidylinositol 4,5-Diphosphate 158-171 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 23793622-4 2013 In this study, we further explored the effect of cholesterol enrichment on HERG channel kinetics. Cholesterol 49-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-79 23793622-5 2013 When membrane cholesterol level was mildly increased in human embryonic kidney (HEK) 293 cells expressing HERG channel, the inactivation and deactivation kinetics of HERG current were not affected, but the activation rate was significantly decelerated at all voltages tested. Cholesterol 14-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 23793622-5 2013 When membrane cholesterol level was mildly increased in human embryonic kidney (HEK) 293 cells expressing HERG channel, the inactivation and deactivation kinetics of HERG current were not affected, but the activation rate was significantly decelerated at all voltages tested. Cholesterol 14-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 166-170 23793622-7 2013 These results indicate that the effect of cholesterol enrichment on HERG channel is due to the depletion of PtdIns(4,5)P2. Cholesterol 42-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 23793622-7 2013 These results indicate that the effect of cholesterol enrichment on HERG channel is due to the depletion of PtdIns(4,5)P2. Phosphatidylinositol 4,5-Diphosphate 108-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 23793622-9 2013 Since the effects of cholesterol enrichment on HERG channel were prevented by inhibiting transcription or by inhibiting PLCbeta1 expression, we conclude that increased PLCbeta1 expression leads to the deceleration of HERG channel activation rate via downregulation of PtdIns(4,5)P2. Cholesterol 21-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 23840331-3 2013 It is reported that PD-118057 and thapsigargin can rescue LQT2 without hERG channel blockade, but the precise mechanism of action is unknown. 2-(4-(2-(3,4-dichlorophenyl)ethyl)phenylamino)benzoic acid 20-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 23840331-3 2013 It is reported that PD-118057 and thapsigargin can rescue LQT2 without hERG channel blockade, but the precise mechanism of action is unknown. Thapsigargin 34-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 23840331-5 2013 OBJECTIVE: IN THIS STUDY, WE INVESTIGATED: (a) the effect of PD-118057 and thapsigargin on the current amplitudes of WT-hERG and WT/E637K-hERG channels; (b) the effect of PD-118057 and thapsigargin on the biophysical properties of WT-hERG and WT/E637K-hERG channels; (c) whether drug treatment can rescue channel processing and trafficking defects of the WT/E637K-hERG mutant. Thapsigargin 75-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 138-142 23840331-9 2013 Additionally, thapsigargin shows a similar result as PD-118057 for the WT-hERG channel, but with the exception of attenuating steady-state inactivation. Thapsigargin 14-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 23793622-9 2013 Since the effects of cholesterol enrichment on HERG channel were prevented by inhibiting transcription or by inhibiting PLCbeta1 expression, we conclude that increased PLCbeta1 expression leads to the deceleration of HERG channel activation rate via downregulation of PtdIns(4,5)P2. Cholesterol 21-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 217-221 23793622-9 2013 Since the effects of cholesterol enrichment on HERG channel were prevented by inhibiting transcription or by inhibiting PLCbeta1 expression, we conclude that increased PLCbeta1 expression leads to the deceleration of HERG channel activation rate via downregulation of PtdIns(4,5)P2. Phosphatidylinositol 4,5-Diphosphate 268-281 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 23793622-10 2013 These results confirm a crosstalk between two plasma membrane-enriched lipids, cholesterol and PtdIns(4,5)P2, in the regulation of HERG channels. Cholesterol 79-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 23793622-10 2013 These results confirm a crosstalk between two plasma membrane-enriched lipids, cholesterol and PtdIns(4,5)P2, in the regulation of HERG channels. Phosphatidylinositol 4,5-Diphosphate 95-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 22564166-0 2013 QSAR and pharmacophore analysis of a series of piperidinyl urea derivatives as HERG blockers and H3 antagonists. piperidinyl urea 47-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 23744352-6 2013 Application of the hERG1 activator, the diphenylurea derivative NS1643, inhibits cell proliferation irreversibly. Carbanilides 40-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-24 23744352-6 2013 Application of the hERG1 activator, the diphenylurea derivative NS1643, inhibits cell proliferation irreversibly. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 64-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-24 23418776-2 2013 In the present study we analysed, using a site-directed cysteine and disulfide chemistry approach, whether the eag/PAS (Per/Arnt/Sim) and proximal domains at the HERG N-terminus exert a role in controlling the access of the N-terminal flexible tail to its binding site in the channel core for interaction with the gating machinery. Protactinium 115-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 23418776-4 2013 The state-dependent formation of a disulfide bridge between Cys3 and an endogenous cysteine residue at position 723 in the C-terminal C-linker suggests that the N-terminal tail of HERG can also get into close proximity with the C-linker structures located at the bottom of helix S6. Disulfides 35-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 23418776-4 2013 The state-dependent formation of a disulfide bridge between Cys3 and an endogenous cysteine residue at position 723 in the C-terminal C-linker suggests that the N-terminal tail of HERG can also get into close proximity with the C-linker structures located at the bottom of helix S6. Cysteine 83-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 23917377-0 2013 ZC88, a novel 4-amino piperidine analog, inhibits the growth of neuroblastoma cells through blocking hERG potassium channel. zc88 0-4 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 23917377-0 2013 ZC88, a novel 4-amino piperidine analog, inhibits the growth of neuroblastoma cells through blocking hERG potassium channel. 4-aminopiperidine 14-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 23917377-4 2013 The results showed that ZC88 could block hERG1 and hERG1b channels expressed in Xenopus oocytes in a concentration-dependent manner. zc88 24-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-46 23546015-0 2013 Mechanistic basis for type 2 long QT syndrome caused by KCNH2 mutations that disrupt conserved arginine residues in the voltage sensor. Arginine 95-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-61 23546015-4 2013 Kv11.1 contains several conserved basic amino acids in the fourth transmembrane segment (S4) of the voltage sensor that are important for normal channel trafficking and gating. Amino Acids, Basic 34-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-6 23546015-5 2013 This study sought to determine the mechanism(s) by which LQT2 mutations at conserved arginine residues in S4 (R531Q, R531W or R534L) alter Kv11.1 function. Arginine 85-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 23546015-5 2013 This study sought to determine the mechanism(s) by which LQT2 mutations at conserved arginine residues in S4 (R531Q, R531W or R534L) alter Kv11.1 function. Arginine 85-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-145 23613831-2 2013 Some pathways driven by angiotensin II, nitric oxide and adrenergic receptors blocker are involved in modulating the properties of KCNH2 potassium channels. Nitric Oxide 40-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-136 23613831-7 2013 RESULTS: There were statistically significant interactions between KCNH2 (1956, C>T) polymorphism and DBP change (P = 0.010), MAP change (P = 0.014) on azelnidipine or nitrendipine therapy patients at the end of 6 weeks. azelnidipine 155-167 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-72 23613831-7 2013 RESULTS: There were statistically significant interactions between KCNH2 (1956, C>T) polymorphism and DBP change (P = 0.010), MAP change (P = 0.014) on azelnidipine or nitrendipine therapy patients at the end of 6 weeks. Nitrendipine 171-183 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-72 23712551-7 2013 Low pH similarly reduces Mg(2+) sensitivity of Kv10.1, and we found that the pH sensitivity of Kv11.1 was greatly attenuated at 1 mM Ca(2+). magnesium ion 25-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 95-101 23917959-6 2013 To generate disease model for LQT2 by iPS cells, we should firstly generate iPS cells from the patient with LQT2 and confirm the genomic mutation in iPS cells. IPS 38-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 23917959-6 2013 To generate disease model for LQT2 by iPS cells, we should firstly generate iPS cells from the patient with LQT2 and confirm the genomic mutation in iPS cells. IPS 76-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 23917959-6 2013 To generate disease model for LQT2 by iPS cells, we should firstly generate iPS cells from the patient with LQT2 and confirm the genomic mutation in iPS cells. IPS 76-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 23917959-6 2013 To generate disease model for LQT2 by iPS cells, we should firstly generate iPS cells from the patient with LQT2 and confirm the genomic mutation in iPS cells. IPS 76-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 23917959-6 2013 To generate disease model for LQT2 by iPS cells, we should firstly generate iPS cells from the patient with LQT2 and confirm the genomic mutation in iPS cells. IPS 76-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 23917959-7 2013 In this study, we showed the successful generation of iPS cells from a patient with KCNH2 G603D mutation. IPS 54-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-89 23917959-9 2013 We also confirmed that the KCNH2 G603D (G1808A) mutation was taken over in patient specific iPS cells. IPS 92-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-32 22949429-6 2012 Collectively, when at least 3 of the 4 tools agreed on the pathogenic status of C-terminal nsSNVs located outside the KCNH2/Kv11.1 cyclic nucleotide-binding domain, the topology-specific estimated predictive value improved from 56% to 91%. Nucleotides, Cyclic 131-148 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-130 23103450-4 2013 Arsenic trioxide (As(2)O(3)), which is used to treat acute promyelocytic leukemia, can cause LQTS type 2 (LQT2) by reducing the hERG current through the diversion of hERG trafficking to the cytoplasmic membrane. Arsenic Trioxide 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-104 23103450-4 2013 Arsenic trioxide (As(2)O(3)), which is used to treat acute promyelocytic leukemia, can cause LQTS type 2 (LQT2) by reducing the hERG current through the diversion of hERG trafficking to the cytoplasmic membrane. Arsenic Trioxide 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 23103450-4 2013 Arsenic trioxide (As(2)O(3)), which is used to treat acute promyelocytic leukemia, can cause LQTS type 2 (LQT2) by reducing the hERG current through the diversion of hERG trafficking to the cytoplasmic membrane. Arsenic 18-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-104 23103450-4 2013 Arsenic trioxide (As(2)O(3)), which is used to treat acute promyelocytic leukemia, can cause LQTS type 2 (LQT2) by reducing the hERG current through the diversion of hERG trafficking to the cytoplasmic membrane. Arsenic 18-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 23103450-4 2013 Arsenic trioxide (As(2)O(3)), which is used to treat acute promyelocytic leukemia, can cause LQTS type 2 (LQT2) by reducing the hERG current through the diversion of hERG trafficking to the cytoplasmic membrane. (2)o(3) 20-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-104 23103450-4 2013 Arsenic trioxide (As(2)O(3)), which is used to treat acute promyelocytic leukemia, can cause LQTS type 2 (LQT2) by reducing the hERG current through the diversion of hERG trafficking to the cytoplasmic membrane. (2)o(3) 20-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 23103450-11 2013 Therefore, matrine and oxymatrine may have the potential to cure LQT2 as a potassium channel activator by promoting hERG channel activation and increasing hERG channel expression. oxymatrine 23-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 23672049-12 2013 Liensinine improves protein expression of HERG channe in HERG-HEK cells. liensinine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 23221912-0 2013 Role of the activation gate in determining the extracellular potassium dependency of block of HERG by trapped drugs. Potassium 61-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 23221912-4 2013 Extracellular potassium influences HERG channel inactivation and can alter block of HERG by some drugs. Potassium 14-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 23221912-4 2013 Extracellular potassium influences HERG channel inactivation and can alter block of HERG by some drugs. Potassium 14-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 23221912-6 2013 In this study, we show that block of WT HERG by bepridil and terfenadine, two drugs previously shown to be trapped inside the HERG channel after the channel closes, is insensitive to extracellular potassium over the range of 0 mM to 20 mM. Bepridil 48-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 23221912-6 2013 In this study, we show that block of WT HERG by bepridil and terfenadine, two drugs previously shown to be trapped inside the HERG channel after the channel closes, is insensitive to extracellular potassium over the range of 0 mM to 20 mM. Bepridil 48-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 126-130 23221912-6 2013 In this study, we show that block of WT HERG by bepridil and terfenadine, two drugs previously shown to be trapped inside the HERG channel after the channel closes, is insensitive to extracellular potassium over the range of 0 mM to 20 mM. Terfenadine 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 23221912-6 2013 In this study, we show that block of WT HERG by bepridil and terfenadine, two drugs previously shown to be trapped inside the HERG channel after the channel closes, is insensitive to extracellular potassium over the range of 0 mM to 20 mM. Terfenadine 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 126-130 23221912-7 2013 We also show that bepridil block of the HERG mutant D540K, a mutant channel that is unable to trap drugs, is dependent on extracellular potassium, correlates with the permeant ion, and is independent of HERG inactivation. Bepridil 18-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 23221912-7 2013 We also show that bepridil block of the HERG mutant D540K, a mutant channel that is unable to trap drugs, is dependent on extracellular potassium, correlates with the permeant ion, and is independent of HERG inactivation. Potassium 136-145 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 23221912-8 2013 These results suggest that the lack of extracellular potassium dependency of block of HERG by some drugs may in part be related to the ability of these drugs to be trapped inside the channel after the channel closes. Potassium 53-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 24069049-14 2013 GA significantly inhibited the potassium currents in a dose- and voltage-dependent manner, suggesting that it exerts its antiarrhythmic action through the prolongation of APD and ERP owing to the inhibition of I K (I Kr, I Ks) and HERG K(+) channel. Glycyrrhetinic Acid 0-2 potassium voltage-gated channel subfamily H member 2 Homo sapiens 231-235 24069049-14 2013 GA significantly inhibited the potassium currents in a dose- and voltage-dependent manner, suggesting that it exerts its antiarrhythmic action through the prolongation of APD and ERP owing to the inhibition of I K (I Kr, I Ks) and HERG K(+) channel. Potassium 31-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 231-235 23358696-3 2013 For instance, ion channels such as the voltage-gated sodium channel, L-type calcium channel, Kv2.1, Kv1.5, Kv4.3 and HERG potassium channel were all reported to be inhibited by CXB. Celecoxib 177-180 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 23672049-0 2013 [Effect of berberine, liensinine and neferine on HERG channel expression]. Berberine 11-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 23672049-0 2013 [Effect of berberine, liensinine and neferine on HERG channel expression]. liensinine 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 23672049-0 2013 [Effect of berberine, liensinine and neferine on HERG channel expression]. neferine 37-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. Berberine 157-166 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. liensinine 168-178 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. neferine 183-191 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. Berberine 330-339 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. Berberine 330-339 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. liensinine 474-484 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-1 2013 OBJECTIVE: Immunofluorescence and Western blot methods were adopted for qualitative and quantitative detections of the effect of different concentrations of berberine, liensinine and neferine on the expression of stable transfection in HERG potassium channel in HEK-293 cells, as well as the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues, in order to investigate the anti-arrhythmic mechanism of berberine, liensinine and neferine. neferine 489-497 potassium voltage-gated channel subfamily H member 2 Homo sapiens 236-240 23672049-3 2013 Immunofluorescence method as well as confocal laser microscope were used to detect the effect of different concentrations of berberine, liensinine and neferine on protein expression of HERG channel. Berberine 125-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 23672049-3 2013 Immunofluorescence method as well as confocal laser microscope were used to detect the effect of different concentrations of berberine, liensinine and neferine on protein expression of HERG channel. liensinine 136-146 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 23672049-3 2013 Immunofluorescence method as well as confocal laser microscope were used to detect the effect of different concentrations of berberine, liensinine and neferine on protein expression of HERG channel. neferine 151-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 23672049-4 2013 Western blot method was used to detect the effect of different concentrations of berberine on protein expression of Ikr channel in cardiac muscular tissues as well as the effect of berberine, liensinine and neferine on protein expression of stable transfection in HERG potassium channel in HEK-293 cells. neferine 207-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 264-268 23672049-6 2013 Berberine (10, 30 micromol x L(-1)) remarkably inhibited protein expression of HERG channel in HERG-HEK cells (P < 0.01). Berberine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 23672049-6 2013 Berberine (10, 30 micromol x L(-1)) remarkably inhibited protein expression of HERG channel in HERG-HEK cells (P < 0.01). Berberine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 95-99 23672049-8 2013 Liensinine (3, 10, 30 micromol x L(-1)) increased protein expression of HERG channel in HERG-HEK cells (P < 0.05). liensinine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 23672049-8 2013 Liensinine (3, 10, 30 micromol x L(-1)) increased protein expression of HERG channel in HERG-HEK cells (P < 0.05). liensinine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 23672049-11 2013 Berberine shows inhibitory effect on protein expressions of in vitro HERG-HEK cells and Ikr channel in rat ventricular tissues. Berberine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 23672049-12 2013 Liensinine improves protein expression of HERG channe in HERG-HEK cells. liensinine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 22989185-1 2012 The human ether-a-go-go related gene 1 (hERG1) K ion channel is a key element for the rapid component of the delayed rectified potassium current in cardiac myocytes. Potassium 127-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-45 22974355-0 2012 Effect of daurisoline on HERG channel electrophysiological function and protein expression. daurisoline 10-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 22889737-10 2012 The HERG blocker E-4031 decreased migration. E 4031 17-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 22856456-9 2012 Collectively, these data suggest that the AZAs physically block the K(+) conductance pathway of hERG1 channels by occluding the cytoplasmic mouth of the open pore. Azaserine 42-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-101 22853924-8 2012 Finally, in vivo administration of LY364947, a pharmacological antagonist of TGF-beta signalling, dramatically prevented interstitial fibrosis and LQTS and abolished aberrant expression of TGF-beta1, HERG, and Kir2.1 in ATO-treated guinea pigs. Ly-364947 35-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-204 23054723-0 2012 Blockade of human HERG K+ channels by rosiglitazone, an antidiabetic drug. Rosiglitazone 38-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 23054723-1 2012 This study examined the effect of rosiglitazone, an oral antidiabetic drug, on human ether-a-gogo-related gene (HERG) channels expressed in human embryonic kidney (HEK293) cells. Rosiglitazone 34-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-116 23054723-3 2012 Rosiglitazone inhibited HERG channels in a concentration-dependent manner, with an IC50 value of 18.8 muM and a Hill coefficient of 1.0. Rosiglitazone 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 23054723-5 2012 The rosiglitazone-induced inhibition of HERG channels was voltagedependent, with a steep increase in inhibition over the voltage range of channel opening. Rosiglitazone 4-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 23054723-8 2012 The present study suggests that rosiglitazone blocks HERG channels by binding to activated and inactivated channels, and rosiglitazone use should thus be carefully monitored in patients with pre-existing QT prolongation. Rosiglitazone 32-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 53-57 22161019-0 2012 Imatinib has the potential to exert its antileukemia effects by down-regulating hERG1 K+ channels in chronic myelogenous leukemia. Imatinib Mesylate 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-85 22161019-4 2012 The present study explored a possible regulatory effect of imatinib upon hERG1 K(+) channels as a means to uncover new molecular events involved in the antileukemic activity of this PTK inhibitor in CML. Imatinib Mesylate 59-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-78 22161019-5 2012 The results demonstrated that hERG1 was highly detected in K562 cells and primary CML cells, and down-regulated by imatinib at mRNA and protein levels. Imatinib Mesylate 115-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-35 22161019-7 2012 Moreover, these antileukemia effects of imatinib were potentiated by E-4031, a specific hERG1 inhibitor. Imatinib Mesylate 40-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-93 22161019-7 2012 Moreover, these antileukemia effects of imatinib were potentiated by E-4031, a specific hERG1 inhibitor. E 4031 69-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-93 22161019-8 2012 Together, these results provide evidence of a novel potential molecular mechanism of antileukemic activities by imatinib which, independent of targeting tyrosine kinase, highlight hERG1 K(+) channels as a therapeutic target for CML treatment. Imatinib Mesylate 112-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-185 22522181-0 2012 HERG potassium channel regulation by the N-terminal eag domain. Methyl 2-acetamido-2-deoxy-beta-D-glucopyranoside 52-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 22573844-4 2012 In silico docking of the activator 1,3-bis-(2-hydroxy-5-trifluoromethylphenyl)-urea (NS1643) to an S1-S6 transmembrane homology model of hERG1 predicted putative binding sites. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 35-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-142 22573844-11 2012 The inward conductance elicited by hyperpolarization of D540K hERG1 was abrogated by NS1643 treatment, suggesting that it alters the inward movement of the S4 segment. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 85-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-67 22292854-10 2012 Importantly, hERG1 K(+) channel inhibitor E-4031 impaired these effects. E 4031 42-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-18 22100990-0 2012 Transfection by eukaryotic expression vector pcDNA3-HERG inhibits the cultured neonatal rabbit ventricular myocyte hypertrophy induced by phenylephrine. Phenylephrine 138-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 22100990-9 2012 HERG overexpression could accelerate repolarization and shorten APD at 90% repolarization prolonged by phenylephrine and partially inhibit myocyte hypertrophy and calcineurin activation. Phenylephrine 103-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 22100990-11 2012 HERG overexpression could enhance the repolarization and inhibit the calcineurin activation and myocyte hypertrophy induced by phenylephrine. Phenylephrine 127-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 22465688-0 2012 Iloperidone (Fanapt ), a novel atypical antipsychotic, is a potent HERG blocker and delays cardiac ventricular repolarization at clinically relevant concentration. iloperidone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 22460808-3 2012 HERG1 protein expression was evaluated by immunohistochemistry in paraffin-embedded tissue specimens from 133 patients with laryngeal/hypopharyngeal squamous cell carcinomas and 75 patients with laryngeal dysplasia, and correlated with clinical data. Paraffin 66-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-5 22465688-0 2012 Iloperidone (Fanapt ), a novel atypical antipsychotic, is a potent HERG blocker and delays cardiac ventricular repolarization at clinically relevant concentration. iloperidone 13-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 22465688-3 2012 (1) In vitro level: whole-cell patch-clamp experiments were performed on HERG-transfected HEK293 cells exposed to iloperidone 0.01-1 mumol/L (n = 35 cells, total) to assess drug effect on HERG current. iloperidone 114-125 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 22465688-10 2012 Iloperidone prolongs the QT interval, the cardiac action potentials and is a potent HERG blocker. iloperidone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 22609834-1 2012 AIM: To compare the effects of two stereoisomeric forms of glycyrrhetinic acid on different components of Na(+) current, HERG and Kv1.5 channel currents. Glycyrrhetinic Acid 59-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 22052944-8 2012 Additionally, LQT2-derived cardiac cells were more sensitive than controls to potentially arrhythmogenic drugs, including sotalol, and demonstrated arrhythmogenic electrical activity. Sotalol 122-129 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 21920426-0 2012 Inhibitory effects of a bisbenzylisoquinline alkaloid dauricine on HERG potassium channels. bisbenzylisoquinline alkaloid 24-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 21920426-0 2012 Inhibitory effects of a bisbenzylisoquinline alkaloid dauricine on HERG potassium channels. dauricine 54-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 21920426-5 2012 AIM OF THE STUDY: The effects of Dau on HERG channels were investigated. dauricine 33-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 21920426-10 2012 Dau inhibited I(HERG) in the open and inactivated states, but not in the closed states. dauricine 0-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 21920426-12 2012 CONCLUSIONS: Dau inhibits HERG encoded potassium channels and this action might be a molecular mechanism for the previously reported APD prolongation with this drug. dauricine 13-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 22392372-2 2012 Each amino acid residue in the first half of the HERG peptide incorporated deuterons with a higher rate than those in the second half of the peptide, consistent with the nuclear magnetic resonance structure of this peptide, with amino acids 1-10 being a flexible coil, whereas amino acids 11-24 are a stable amphipathic helix. Deuterium 75-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 22381725-4 2012 Many explorations have helped in understanding the physiopathology by showing that opioids, including methadone, cause a blockage of the potassium channels of the gene HERG K+P. Methadone 102-111 potassium voltage-gated channel subfamily H member 2 Homo sapiens 168-172 22183728-8 2012 These different effects of ISO on LQT1 and LQT2 were verified by optical mapping of the whole heart, suggesting that ISO-induced EADs are genotype specific. Isoproterenol 27-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 22183728-8 2012 These different effects of ISO on LQT1 and LQT2 were verified by optical mapping of the whole heart, suggesting that ISO-induced EADs are genotype specific. Isoproterenol 117-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 22075718-6 2012 Inhibition of HERG currents by acute or sustained application of E-4031, a specific ERG channel blocker, depolarized SW2 cells by 10-15 mV. E 4031 65-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 22075718-8 2012 Blockage of HERG channels by E-4031 for up to 72 h did not affect cell proliferation. E 4031 29-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 21934630-3 2012 The purpose of the present study was to investigate the effects of GFA and GFG on the HERG channel and its structure-function relationship. guanfu base G 75-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 21934630-5 2012 RESULTS: GFA and GFG inhibited HERG channel current in concentration-, voltage-, and time-dependent manners. guanfu base G 17-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 21934630-9 2012 CONCLUSIONS: Both GFA and GFG blocked HERG channel current. guanfu base G 26-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 21951015-9 2012 Lastly, we examined a potential role for hypokalemia as a contributory factor to the patient"s lethal arrhythmia by possible low-potassium-induced degradation of WT HERG and haplo-insufficiency of G816V HERG. Potassium 129-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-169 21951015-9 2012 Lastly, we examined a potential role for hypokalemia as a contributory factor to the patient"s lethal arrhythmia by possible low-potassium-induced degradation of WT HERG and haplo-insufficiency of G816V HERG. Potassium 129-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 203-207 22396785-0 2012 Changes in channel trafficking and protein stability caused by LQT2 mutations in the PAS domain of the HERG channel. Aminosalicylic Acid 85-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 22396785-0 2012 Changes in channel trafficking and protein stability caused by LQT2 mutations in the PAS domain of the HERG channel. Aminosalicylic Acid 85-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 22396785-2 2012 Several LQT2-associated mutations have been mapped to the amino terminal cytoplasmic Per-Arnt-Sim (PAS) domain of the HERG1a channel subunit. Aminosalicylic Acid 99-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 8-12 22396785-2 2012 Several LQT2-associated mutations have been mapped to the amino terminal cytoplasmic Per-Arnt-Sim (PAS) domain of the HERG1a channel subunit. Aminosalicylic Acid 99-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-123 22124116-3 2011 Isolated, PAS-containing hERG1a N-terminal regions (NTRs) directly regulate NTR-deleted hERG1a channels; however, it is unclear whether hERG1b isoforms contain sufficient machinery to support regulation by hERG1a NTRs. Aminosalicylic Acid 10-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-30 22020779-2 2012 The aim of this study was to investigate the role of HERG in cisplatin-induced apoptosis in gastric cancer in vitro and in vivo. Cisplatin 73-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 22020779-7 2012 Our results show that cisplatin increased the expression of HERG in gastric cancer cells. Cisplatin 22-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 22020779-8 2012 Silencing HERG inhibited the apoptosis induced by cisplatin both in vitro and in vivo, by attenuating the cisplatin effects on Bcl-2, Bax and active caspase-3. Cisplatin 62-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 10-14 22020779-8 2012 Silencing HERG inhibited the apoptosis induced by cisplatin both in vitro and in vivo, by attenuating the cisplatin effects on Bcl-2, Bax and active caspase-3. Cisplatin 118-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 10-14 22020779-9 2012 The role of HERG in modulation of cisplatin-induced apoptosis suggests that HERG may provide a new potential target for cisplatin chemotherapy in human gastric cancer. Cisplatin 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 22020779-9 2012 The role of HERG in modulation of cisplatin-induced apoptosis suggests that HERG may provide a new potential target for cisplatin chemotherapy in human gastric cancer. Cisplatin 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 22020779-9 2012 The role of HERG in modulation of cisplatin-induced apoptosis suggests that HERG may provide a new potential target for cisplatin chemotherapy in human gastric cancer. Cisplatin 132-141 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 22020779-9 2012 The role of HERG in modulation of cisplatin-induced apoptosis suggests that HERG may provide a new potential target for cisplatin chemotherapy in human gastric cancer. Cisplatin 132-141 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 22124116-3 2011 Isolated, PAS-containing hERG1a N-terminal regions (NTRs) directly regulate NTR-deleted hERG1a channels; however, it is unclear whether hERG1b isoforms contain sufficient machinery to support regulation by hERG1a NTRs. Aminosalicylic Acid 10-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-93 22124116-3 2011 Isolated, PAS-containing hERG1a N-terminal regions (NTRs) directly regulate NTR-deleted hERG1a channels; however, it is unclear whether hERG1b isoforms contain sufficient machinery to support regulation by hERG1a NTRs. Aminosalicylic Acid 10-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-93 22124116-10 2011 Our results demonstrate that deactivation is faster in hERG1a/hERG1b channels compared to hERG1a channels because of fewer PAS domains, not because of an inhibitory effect of the unique hERG1b NTR. Aminosalicylic Acid 123-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-60 22124116-10 2011 Our results demonstrate that deactivation is faster in hERG1a/hERG1b channels compared to hERG1a channels because of fewer PAS domains, not because of an inhibitory effect of the unique hERG1b NTR. Aminosalicylic Acid 123-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-67 21839071-6 2011 Characteristics of blockage on hHCN channels were consistent with those of amiodarone as "trapped" drugs on human ether-a-go-go-related gene (HERG) channels. Amiodarone 75-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 22003215-10 2011 Similarly, the incidence of AP prolongations in the presence of 3 mumol/L bupivacaine was significantly increased from 6% in control myocytes to 24% in LQT1-like but not in LQT2-like myocytes. Bupivacaine 74-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 21856740-1 2011 Two different mechanisms leading to increased current have been described for the small-molecule human ether-a-go-go-related gene (herg) activator NS1643 [1,3-bis-(2-hydroxy-5-trifluoromethylphenyl)-urea]. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 147-153 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 22396785-3 2012 Here we have characterized the trafficking properties of some LQT2-associated PAS domain mutants and analyzed rescue of the trafficking mutants by low temperature (27 C) or by the pore blocker drug E4031. Aminosalicylic Acid 78-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 22396785-4 2012 We show that the LQT2-associated mutations in the PAS domain of the HERG channel display molecular properties that are distinct from the properties of LQT2-associated mutations in the trans-membrane region. Aminosalicylic Acid 50-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 22396785-4 2012 We show that the LQT2-associated mutations in the PAS domain of the HERG channel display molecular properties that are distinct from the properties of LQT2-associated mutations in the trans-membrane region. Aminosalicylic Acid 50-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 22396785-5 2012 Unlike the latter, many of the tested PAS domain LQT2-associated mutations do not result in trafficking deficiency of the channel. Aminosalicylic Acid 38-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 21856740-1 2011 Two different mechanisms leading to increased current have been described for the small-molecule human ether-a-go-go-related gene (herg) activator NS1643 [1,3-bis-(2-hydroxy-5-trifluoromethylphenyl)-urea]. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 155-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 22260022-0 2011 [Lidamycin inhibits the proliferation of HERG K+ channel highly expressing cancer cells and shows synergy with anticancer drugs]. C 1027 1-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 22260022-1 2011 This study is to investigate inhibitory effects of lidamycin (LDM) on the proliferation of HERG K+ channel highly expressing cancer cells and its synergy with anticancer drugs. C 1027 51-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 22260022-2 2011 MTT assay was used to examine the inhibitory effects of lidamycin combined with various anticancer drugs on the proliferation of human lung cancer A549 cells, human colon cancer HT-29 cells and herg-stably-transfected A549 cells. monooxyethylene trimethylolpropane tristearate 0-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 194-198 22260022-2 2011 MTT assay was used to examine the inhibitory effects of lidamycin combined with various anticancer drugs on the proliferation of human lung cancer A549 cells, human colon cancer HT-29 cells and herg-stably-transfected A549 cells. C 1027 56-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 194-198 22260022-6 2011 The efficacy in HT-29 cells with high HERG potassium expression level is less potent than that in A549 cells with low expression level. Potassium 43-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 22260022-13 2011 HERG expression level negatively correlated with inhibitory effect on the proliferation of cancer cells by DOX. Doxorubicin 107-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 21798421-7 2011 The average QTc was 314 +- 23 ms. A mutation in genes related to SQTS was found in 23% of the probands; most of them had a gain of function mutation in HERG (SQTS1). qtc 12-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 152-156 22128262-1 2011 The effect of cyclosporin A (CsA), an immunosuppressant, on human ether-a-go-go-related gene (HERG) channel as it is expressed in human embryonic kidney cells was studied using a whole-cell, patch-clamp technique. Cyclosporine 29-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 22128262-2 2011 CsA inhibited the HERG channel in a concentration-dependent manner, with an IC(50) value and a Hill coefficient of 3.17 microM and 0.89, respectively. Cyclosporine 0-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 22128262-4 2011 The CsA-induced inhibition of HERG channels was voltage-dependent, with a steep increase over the voltage range of the channel opening. Cyclosporine 4-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 22128262-6 2011 CsA blocked the HERG channels predominantly in the open and inactivated states rather than in the closed state. Cyclosporine 0-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 22128262-7 2011 Results of the present study suggest that CsA acts directly on the HERG channel as an open-channel blocker, and it acts independently of its effect on calcineurin activity. Cyclosporine 42-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 21743002-4 2011 3-Nitro-n-(4-phenoxyphenyl) benzamide [ICA-105574 (ICA)] has been discovered to activate hERG1 by strong attenuation of pore-type inactivation. 3-nitro-N-(4-phenoxyphenyl)benzamide 0-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-94 21743002-4 2011 3-Nitro-n-(4-phenoxyphenyl) benzamide [ICA-105574 (ICA)] has been discovered to activate hERG1 by strong attenuation of pore-type inactivation. 3-nitro-N-(4-phenoxyphenyl)benzamide 39-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-94 21743002-4 2011 3-Nitro-n-(4-phenoxyphenyl) benzamide [ICA-105574 (ICA)] has been discovered to activate hERG1 by strong attenuation of pore-type inactivation. isocyanic acid 39-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-94 21743002-5 2011 Here, we used scanning mutagenesis of hERG1 to identify the molecular determinants of ICA action. isocyanic acid 86-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-43 21743002-8 2011 This was confirmed by showing that the noninactivating mutant hERG1 channel (G628C/S631C) was inhibited by ICA and that the addition of the F557L mutation rendered the channel drug-insensitive. isocyanic acid 107-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-67 21743002-9 2011 Simulated molecular docking of ICA to homology models of hERG1 corroborated the scanning mutagenesis findings. isocyanic acid 31-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-62 21743002-10 2011 Together, our findings indicate that ICA is a mixed agonist of hERG1 channels. isocyanic acid 37-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-68 21449979-8 2011 In contrast, E-4031 was a more potent blocker of hERG 1a compared with 1a/1b channels, as previously reported, as was dofetilide, another high-affinity blocker. E 4031 13-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-55 20547773-6 2011 The results showed that drugs in group 1 induced greater inhibition of human ether-a-go-go-related gene (HERG) potassium current or the rapid component of the delayed rectifier potassium current (I(kr)), had lower half-maximal inhibitory concentration (IC50) values for inhibition of HERG/I(kr), and induced greater QTc increases in humans. qtc 316-319 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 21490315-5 2011 We found that treating cells in nocodazole, a microtubule depolymerizing agent, altered the subcellular localization, functional expression, and glycosylation of the LQT2 mutation G601S-hERG differently from wild-type hERG (WT-hERG). Nocodazole 32-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 166-170 21497594-10 2011 Thus, we conclude that CVB-D inhibits HERG encoded potassium channels and this action might be a molecular mechanism for the previously reported APD prolongation and QT interval prolongation with this drug. PVB protocol 23-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 21525004-0 2011 Progesterone impairs human ether-a-go-go-related gene (HERG) trafficking by disruption of intracellular cholesterol homeostasis. Cholesterol 104-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 21525004-1 2011 The prolongation of QT intervals in both mothers and fetuses during the later period of pregnancy implies that higher levels of progesterone may regulate the function of the human ether-a-go-go-related gene (HERG) potassium channel, a key ion channel responsible for controlling the length of QT intervals. Progesterone 128-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 208-212 21525004-3 2011 Treatment with progesterone for 24 h decreased the abundance of the fully glycosylated form of the HERG channel in rat neonatal cardiac myocytes and HERG-HEK293 cells, a cell line stably expressing HERG channels. Progesterone 15-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 21525004-3 2011 Treatment with progesterone for 24 h decreased the abundance of the fully glycosylated form of the HERG channel in rat neonatal cardiac myocytes and HERG-HEK293 cells, a cell line stably expressing HERG channels. Progesterone 15-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 21525004-3 2011 Treatment with progesterone for 24 h decreased the abundance of the fully glycosylated form of the HERG channel in rat neonatal cardiac myocytes and HERG-HEK293 cells, a cell line stably expressing HERG channels. Progesterone 15-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 21525004-4 2011 Progesterone also concentration-dependently decreased HERG current density, but had no effect on voltage-gated L-type Ca(2+) and K(+) channels. Progesterone 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 21525004-5 2011 Immunofluorescence microscopy and Western blot analysis show that progesterone preferentially decreased HERG channel protein abundance in the plasma membrane, induced protein accumulation in the dilated endoplasmic reticulum (ER), and increased the protein expression of C/EBP homologous protein, a hallmark of ER stress. Progesterone 66-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 21525004-6 2011 Application of 2-hydroxypropyl-beta-cyclodextrin (a sterol-binding agent) or overexpression of Rab9 rescued the progesterone-induced HERG trafficking defect and ER stress. 2-Hydroxypropyl-beta-cyclodextrin 15-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 21525004-6 2011 Application of 2-hydroxypropyl-beta-cyclodextrin (a sterol-binding agent) or overexpression of Rab9 rescued the progesterone-induced HERG trafficking defect and ER stress. Sterols 52-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 21525004-6 2011 Application of 2-hydroxypropyl-beta-cyclodextrin (a sterol-binding agent) or overexpression of Rab9 rescued the progesterone-induced HERG trafficking defect and ER stress. Progesterone 112-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 21525004-7 2011 Disruption of intracellular cholesterol homeostasis with simvastatin, imipramine, or exogenous application of cholesterol mimicked the effect of progesterone on HERG channel trafficking. Cholesterol 28-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 161-165 21525004-7 2011 Disruption of intracellular cholesterol homeostasis with simvastatin, imipramine, or exogenous application of cholesterol mimicked the effect of progesterone on HERG channel trafficking. Cholesterol 110-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 161-165 21525004-7 2011 Disruption of intracellular cholesterol homeostasis with simvastatin, imipramine, or exogenous application of cholesterol mimicked the effect of progesterone on HERG channel trafficking. Progesterone 145-157 potassium voltage-gated channel subfamily H member 2 Homo sapiens 161-165 21536673-3 2011 Only a small percentage of hERG channels containing PAS domain LQT2 mutations (hERG PAS-LQT2) have been characterized in mammalian cells, so the functional effect of these mutations is unclear. Aminosalicylic Acid 52-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 21536673-3 2011 Only a small percentage of hERG channels containing PAS domain LQT2 mutations (hERG PAS-LQT2) have been characterized in mammalian cells, so the functional effect of these mutations is unclear. Aminosalicylic Acid 52-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 21536673-11 2011 Thus, our results reveal a putative "gating face" in the PAS domain where mutations within this region form functional channels with altered gating properties, and we show that NPAS is a general means for rescuing aberrant gating in hERG LQT2 mutant channels and may be a potential biological therapeutic. Aminosalicylic Acid 57-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 238-242 21536683-8 2011 Rerouting this inhibitor to the outer mitochondrial membrane diminishes its ability to block cAMP-dependent HERG induction. Cyclic AMP 93-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 21776218-7 2011 Expression of the ion-channel genes CACNA1, KCNH2, KCNQ1 and KCNE1 were down-regulated by 1-muM arsenic. Arsenic 96-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-49 21394035-7 2011 RESULTS: (1) The estimated concentration at which 50% of the maximal inhibitory effect is observed (IC(50)) for paliperidone on HERG current was 0.5276 mumol/L. Paliperidone Palmitate 112-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 21394035-11 2011 CONCLUSIONS: Paliperidone prolongs the QT interval by blocking HERG current at clinically relevant concentrations and is potentially unsafe. Paliperidone Palmitate 13-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 23717061-0 2011 Differential effects of ginsenoside metabolites on HERG k channel currents. Ginsenosides 24-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 23717061-3 2011 In a previous report we demonstrated that ginsenoside Rg3 regulates HERG K(+) channels by decelerating deactivation. Ginsenosides 42-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 23717061-4 2011 However, little is known about how ginsenoside metabolites regulate HERG K(+) channel activity. Ginsenosides 35-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 23717061-5 2011 In the present study, we examined the effects of ginsenoside metabolites such as compound K (CK), protopanaxadiol (PPD), and protopanaxatriol (PPT) on HERG K(+) channel activity by expressing human alpha subunits in Xenopus oocytes. Ginsenosides 49-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-155 23717061-5 2011 In the present study, we examined the effects of ginsenoside metabolites such as compound K (CK), protopanaxadiol (PPD), and protopanaxatriol (PPT) on HERG K(+) channel activity by expressing human alpha subunits in Xenopus oocytes. protopanaxadiol 98-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-155 23717061-5 2011 In the present study, we examined the effects of ginsenoside metabolites such as compound K (CK), protopanaxadiol (PPD), and protopanaxatriol (PPT) on HERG K(+) channel activity by expressing human alpha subunits in Xenopus oocytes. protopanaxatriol 125-141 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-155 23717061-5 2011 In the present study, we examined the effects of ginsenoside metabolites such as compound K (CK), protopanaxadiol (PPD), and protopanaxatriol (PPT) on HERG K(+) channel activity by expressing human alpha subunits in Xenopus oocytes. protopanaxatriol 143-146 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-155 23717061-10 2011 These results indicate that ginsenoside metabolites exhibit differential regulation on Ideactivating-tail of HERG K(+) channel. Ginsenosides 28-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 21175572-0 2011 Trapping and dissociation of propafenone derivatives in HERG channels. Propafenone 29-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 22977528-0 2011 Arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells through activation of caspase-3 and inhibition of HERG channels. Arsenic Trioxide 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 22977528-8 2011 Taken together, the results indicate that arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells at least in part through the activation of caspase-3 and the decrease in HERG expression. Arsenic Trioxide 42-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 189-193 21496174-0 2011 Patient with syncope and LQTS carrying a mutation in the PAS domain of the hERG1 channel. Protactinium 57-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-80 21496174-6 2011 Identifying mutations in the PAS domain or other domains of the hERG1 channel and understanding their effect may provide more focused and mutation-specific risk assessment in this population. Protactinium 29-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-69 21175572-3 2011 A small molecule-like propafenone is efficiently trapped in the closed HERG channel conformation. Propafenone 22-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 21175572-12 2011 CONCLUSION AND IMPLICATIONS: The data show that extending the size of a trapped HERG blocker-like propafenone by adding a bulky side chain may impede channel closure and thereby facilitate drug dissociation at rest. Propafenone 98-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 21367833-5 2011 When LQT2-hiPSC cardiomyocytes were exposed to E4031 (an I(Kr) blocker), arrhythmias developed and these presented as early after depolarizations (EADs) in the action potentials. E 4031 47-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 5-9 21367833-6 2011 In contrast to control cardiomyocytes, LQT2-hiPSC cardiomyocytes also developed EADs when challenged with the clinically used stressor, isoprenaline. Isoproterenol 136-148 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 21109023-9 2011 Moreover, the results obtained demonstrate the importance of the glutamic acid at position 637 for the inactivation process and K(+) selectivity of Kv11.1 channels. Glutamic Acid 65-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-154 21063774-10 2011 Genetic polymorphism in KCNH2 was associated with effectiveness of domperidone (p=0.041). Domperidone 67-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-29 21278233-3 2011 Although diverse compounds reduce the hERG current (I(hERG)) by blocking the channel, probucol, a cholesterol-lowering drug that causes LQTS, reduces I(hERG) by decreasing plasma-membrane hERG protein expression. Probucol 86-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-59 21130771-10 2011 Both quinidine and sotalol may be therapeutic options for patients with the T618I HERG mutation. Quinidine 5-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 21130771-10 2011 Both quinidine and sotalol may be therapeutic options for patients with the T618I HERG mutation. Sotalol 19-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 22156660-3 2011 Fluoroquinolones prolong the QT interval by blocking voltage-gated potassium channels, especially the rapid component of the delayed rectifier potassium current I(Kr), expressed by HERG (the human ether-a-go-go-related gene). Fluoroquinolones 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-185 21063774-13 2011 CONCLUSIONS: Genetic characteristics associated with response to domperidone therapy included polymorphisms in the drug transporter gene ABCB1, the potassium channel KCNH2 gene, and alpha1D--adrenoceptor ADRA1D gene. Domperidone 65-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 166-171 20876384-0 2011 Molecular determinants of human ether-a-go-go-related gene 1 (hERG1) K+ channel activation by NS1643. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 94-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-67 20876384-5 2011 We defined previously the mechanism of action of 1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea (NS1643), a compound that increases hERG1 currents by shifting the voltage-dependence of inactivation to more positive potentials. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 49-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 135-140 20876384-5 2011 We defined previously the mechanism of action of 1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea (NS1643), a compound that increases hERG1 currents by shifting the voltage-dependence of inactivation to more positive potentials. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 100-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 135-140 20876384-8 2011 Some of these residues form a cluster and, together with molecular modeling, suggest that NS1643 binds to a pocket near the extracellular ends of the S5/S6 segments of two adjacent hERG1 channel subunits. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 90-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-186 22039467-0 2011 Inhibition of HERG potassium channels by celecoxib and its mechanism. Celecoxib 41-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 21351488-0 2010 [Chinfloxacin hydrochloride inhibits HERG potassium channel at open state]. chinfloxacin hydrochloride 1-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 21351488-1 2010 This study is designed to investigate the effects of chinfloxacin hydrochloride (CFX) on the kinetics of HERG K+ channel. chinfloxacin hydrochloride 53-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 21351488-1 2010 This study is designed to investigate the effects of chinfloxacin hydrochloride (CFX) on the kinetics of HERG K+ channel. Cefoxitin 81-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 20693282-6 2010 We identified an intrinsically weak, noncanonical poly(A) signal, AGUAAA, within intron 9 of hERG1 that modulates the expression of hERG1a and hERG1a(USO). Poly A 50-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-98 21035456-7 2011 The restoration of functional expression by antisense morpholino oligonucleotides was also observed in LQT2 frameshift mutations. Morpholinos 54-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 21035456-8 2011 Our findings suggest that inhibition of NMD by antisense morpholino oligonucleotides may be a potential therapeutic approach for some LQT2 patients carrying nonsense and frameshift mutations. Morpholinos 57-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 134-138 20674746-3 2010 The present study investigated whether raloxifene could affect the cloned hERG channel (I(hERG)) and recombinant human cardiac KCNQ1/KCNE1 channel (I(Ks)) stably expressed in HEK 293 cells using a patch-clamp technique. Raloxifene Hydrochloride 39-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-95 20693282-8 2010 In contrast, eliminating the intron 9 poly(A) signal or increasing the strength of 5" splice site led to the predominant production of hERG1a and a significant increase in hERG1 current. Poly A 38-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 135-140 20601449-5 2010 LQT1-like behavior was pharmacologically induced by chromanol 293B (10 micromol/L) and LQT2-like states by E4031 (10 micromol/L). E 4031 107-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 20535583-1 2010 OBJECTIVE: To observe the effect of matrine on human ether a go-go related gene (HERG) potassium channels expressed in Chinese hamster ovary (CHO) cells and investigate whether HERG channel is a new target of the pharmacological effect of matrine on arrhythmia and tumor METHODS: HERG channel potassium current in CHO cell was recorded using whole-cell patch-clamp technique, and the influence of matrine on the current was explored. matrine 36-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 20535583-2 2010 RESULTS: Matrine inhibited HERG potassium current in a dose-dependent manner, and the 50% inhibitory concentration (IC IC(50)) was 411+-23 mumol/L. Potassium 32-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 20535583-3 2010 Matrine had no significant effect on the activation kinetics, and mainly blocked HERG channels in their closed state. matrine 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 21165329-0 2010 Response of i(kr) and HERG currents to the antipsychotics tiapride and sulpiride. Tiapride Hydrochloride 58-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 21165329-0 2010 Response of i(kr) and HERG currents to the antipsychotics tiapride and sulpiride. Sulpiride 71-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 21165329-4 2010 However, our findings did show that tiapride increased the potential for half-maximal activation (V(1/2)) of HERG at 10~300 microM, whereas sulpiride increased the maximum conductance (G(max)) at 3, 10 and 100 microM. Tiapride Hydrochloride 36-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 20734202-1 2010 The goal of the present study was to examine the effects of lobeline, an agonist at nicotinic receptors, on cloned Kv channels, Kv1.5, Kv3.1, Kv4.3, and human ether-a-gogo-related gene (HERG), which are stably expressed in Chinese hamster ovary (CHO) or human embryonic kidney 293 (HEK293) cells. Lobeline 60-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 186-190 20734202-10 2010 Lobeline also inhibited Kv3.1, Kv4.3, and HERG in a concentration-dependent manner, with IC(50) values of 21.7, 28.2, and 0.34 muM, respectively. Lobeline 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 20601449-9 2010 In LQT1-like myocytes, droperidol (0.6 micromol/L) further prolonged action potentials by 31% + or - 6% (n = 6) but shortened action potentials of LQT2-like myocytes by 11% + or - 2% (n = 8). Droperidol 23-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 147-151 20394569-5 2010 In vitro studies demonstrated cardiac electrophysiological effects of domperidone on the rapid component of the cardiac delayed rectifier K(+) current (I(Kr)) through the blockade of channels encoded by the Human Ether-a-go-go Related Gene (HERG). Domperidone 70-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 241-245 20547678-0 2010 Cysteine 723 in the C-linker segment confers oxidative inhibition of hERG1 potassium channels. Cysteine 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-74 20547678-3 2010 To investigate the molecular mechanisms of hERG1 channel alteration by ROS, hERG1 and mutants thereof were expressed in HEK293 cells and studied with the whole-cell patch-clamp method. Reactive Oxygen Species 71-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-48 20662825-0 2010 The action of a novel fluoroquinolone antibiotic agent antofloxacin hydrochloride on human-ether-a-go-go-related gene potassium channel. Fluoroquinolones 22-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-117 20662825-0 2010 The action of a novel fluoroquinolone antibiotic agent antofloxacin hydrochloride on human-ether-a-go-go-related gene potassium channel. antofloxacin 55-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-117 20662825-2 2010 In this study, the effects of a novel fluoroquinolone, antofloxacin hydrochloride (AX) on human-ether-a-go-go-related gene (HERG) encoding potassium channels and the biophysical mechanisms of drug action were performed with whole-cell patch-clamp technique in transiently transfected HEK293 cells. Fluoroquinolones 38-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-122 20662825-2 2010 In this study, the effects of a novel fluoroquinolone, antofloxacin hydrochloride (AX) on human-ether-a-go-go-related gene (HERG) encoding potassium channels and the biophysical mechanisms of drug action were performed with whole-cell patch-clamp technique in transiently transfected HEK293 cells. Fluoroquinolones 38-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 20662825-2 2010 In this study, the effects of a novel fluoroquinolone, antofloxacin hydrochloride (AX) on human-ether-a-go-go-related gene (HERG) encoding potassium channels and the biophysical mechanisms of drug action were performed with whole-cell patch-clamp technique in transiently transfected HEK293 cells. antofloxacin 55-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-122 20662825-2 2010 In this study, the effects of a novel fluoroquinolone, antofloxacin hydrochloride (AX) on human-ether-a-go-go-related gene (HERG) encoding potassium channels and the biophysical mechanisms of drug action were performed with whole-cell patch-clamp technique in transiently transfected HEK293 cells. antofloxacin 55-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 20662825-2 2010 In this study, the effects of a novel fluoroquinolone, antofloxacin hydrochloride (AX) on human-ether-a-go-go-related gene (HERG) encoding potassium channels and the biophysical mechanisms of drug action were performed with whole-cell patch-clamp technique in transiently transfected HEK293 cells. antofloxacin 83-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-122 20662825-2 2010 In this study, the effects of a novel fluoroquinolone, antofloxacin hydrochloride (AX) on human-ether-a-go-go-related gene (HERG) encoding potassium channels and the biophysical mechanisms of drug action were performed with whole-cell patch-clamp technique in transiently transfected HEK293 cells. antofloxacin 83-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 20662825-4 2010 In comparison with sparfloxacin (SPX), levofloxacin lactate (LVFX), the potency of AX to inhibit HERG tail currents was the least one, with an IC(50) value of 460.37 microM. antofloxacin 83-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 20463060-4 2010 Stimulation of alpha1A-AR with phenylephrine or direct activation of PKC with phorbol ester increased HERG channel protein abundance and K(+) current density in a time- and dose-dependent manner. Phenylephrine 31-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 20463060-4 2010 Stimulation of alpha1A-AR with phenylephrine or direct activation of PKC with phorbol ester increased HERG channel protein abundance and K(+) current density in a time- and dose-dependent manner. Phorbol Esters 78-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 20463060-6 2010 Phorbol ester and moderate alpha1A-AR stimulation enhanced HERG abundance in a PKC-dependent fashion but with stronger alpha1A-adrenergic stimulation; protein kinase A (PKA)-dependent activity also contributed. Phorbol Esters 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 20428834-0 2010 HERG K+ channel related chemosensitivity to sparfloxacin in colon cancer cells. sparfloxacin 44-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 20428834-3 2010 The aim of this study was to investigate the effects of antibacterial agents sparfloxacin (SPFX), a blocker of HERG channel, on HERG K+ channel highly expressing colon cancer cells. sparfloxacin 77-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 111-115 20428834-3 2010 The aim of this study was to investigate the effects of antibacterial agents sparfloxacin (SPFX), a blocker of HERG channel, on HERG K+ channel highly expressing colon cancer cells. sparfloxacin 77-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 20428834-3 2010 The aim of this study was to investigate the effects of antibacterial agents sparfloxacin (SPFX), a blocker of HERG channel, on HERG K+ channel highly expressing colon cancer cells. sparfloxacin 91-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 111-115 20428834-3 2010 The aim of this study was to investigate the effects of antibacterial agents sparfloxacin (SPFX), a blocker of HERG channel, on HERG K+ channel highly expressing colon cancer cells. sparfloxacin 91-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 20428834-14 2010 Study with herg-transfected HEK293 cells and siRNA-knock down HCT116 cells confirmed that the cell viability inhibition by SPFX was correlated with HERG expression. sparfloxacin 123-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 11-15 20428834-14 2010 Study with herg-transfected HEK293 cells and siRNA-knock down HCT116 cells confirmed that the cell viability inhibition by SPFX was correlated with HERG expression. sparfloxacin 123-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 20660661-5 2010 In human ether-a-go-go-related gene type 1 (hERG1) channels, Cd(2+) coordinated by D456 and D460 in S2 and D509 in S3 induces a positive shift in the voltage dependence of activation of ionic currents. d456 83-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-49 20660661-5 2010 In human ether-a-go-go-related gene type 1 (hERG1) channels, Cd(2+) coordinated by D456 and D460 in S2 and D509 in S3 induces a positive shift in the voltage dependence of activation of ionic currents. d460 92-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-49 20660661-5 2010 In human ether-a-go-go-related gene type 1 (hERG1) channels, Cd(2+) coordinated by D456 and D460 in S2 and D509 in S3 induces a positive shift in the voltage dependence of activation of ionic currents. d509 107-111 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-49 20660661-11 2010 New Markov models of hERG1 channels were developed that describe gating currents as a noncooperative two-phase process of the VSD and can account for changes in these currents caused by extracellular Cd(2+). Cadmium 200-202 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-26 20375276-0 2010 Ceramide modulates HERG potassium channel gating by translocation into lipid rafts. Ceramides 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 20375276-4 2010 Yet the acute effects of ceramide on HERG potassium channels are not known. Ceramides 25-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 20375276-5 2010 In the present study we examined the effects of cell-permeable C(6)-ceramide on HERG potassium channels stably expressed in HEK-293 cells. N-caproylsphingosine 63-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 20375276-6 2010 C(6)-ceramide (10 microM) reversibly inhibited HERG channel current (I(HERG)) by 36 +/- 5%. N-caproylsphingosine 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 20375276-6 2010 C(6)-ceramide (10 microM) reversibly inhibited HERG channel current (I(HERG)) by 36 +/- 5%. N-caproylsphingosine 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 20375276-8 2010 Mechanistically, ceramide recruited HERG channels within caveolin-enriched lipid rafts. Ceramides 17-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 20375276-11 2010 Our results provide new insights into the effects of C(6)-ceramide on HERG channels and suggest that C(6)-ceramide can be a promising therapeutic for cancers that overexpress HERG. N-caproylsphingosine 53-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 20375276-11 2010 Our results provide new insights into the effects of C(6)-ceramide on HERG channels and suggest that C(6)-ceramide can be a promising therapeutic for cancers that overexpress HERG. N-caproylsphingosine 101-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 20375276-11 2010 Our results provide new insights into the effects of C(6)-ceramide on HERG channels and suggest that C(6)-ceramide can be a promising therapeutic for cancers that overexpress HERG. N-caproylsphingosine 101-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 175-179 20348026-1 2010 BACKGROUND: Mutations in the KCNQ1 and human ether-a-go-go-related gene (HERG) genes cause the long QT syndromes, LQTS1 and LQTS2, due to reductions in the cardiac repolarizing I(Ks) and I(Kr) currents, respectively. Potassium 179-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 20097192-0 2010 The amiodarone derivative KB130015 activates hERG1 potassium channels via a novel mechanism. Amiodarone 4-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-50 20097192-0 2010 The amiodarone derivative KB130015 activates hERG1 potassium channels via a novel mechanism. KB 130015 26-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-50 20097192-2 2010 Since they are targets of various drugs with cardiac side effects we tested whether the amiodarone derivative 2-methyl-3-(3,5-diiodo-4-carboxymethoxybenzyl)benzofuran (KB130015) blocks hERG1 channels like its parent compound. Amiodarone 88-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-190 20097192-2 2010 Since they are targets of various drugs with cardiac side effects we tested whether the amiodarone derivative 2-methyl-3-(3,5-diiodo-4-carboxymethoxybenzyl)benzofuran (KB130015) blocks hERG1 channels like its parent compound. KB 130015 110-166 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-190 20097192-2 2010 Since they are targets of various drugs with cardiac side effects we tested whether the amiodarone derivative 2-methyl-3-(3,5-diiodo-4-carboxymethoxybenzyl)benzofuran (KB130015) blocks hERG1 channels like its parent compound. KB 130015 168-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-190 20097192-6 2010 KB130015 presumably binds to the hERG1 pore from the cytosolic side and functionally competes with hERG1 block by amiodarone, E4031 (N-[4-[[1-[2-(6-methyl-2-pyridinyl)ethyl]-4-piperidinyl] carbonyl] phenyl] methanesulfonamide dihydrochloride), and sertindole. Amiodarone 114-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-104 20097192-6 2010 KB130015 presumably binds to the hERG1 pore from the cytosolic side and functionally competes with hERG1 block by amiodarone, E4031 (N-[4-[[1-[2-(6-methyl-2-pyridinyl)ethyl]-4-piperidinyl] carbonyl] phenyl] methanesulfonamide dihydrochloride), and sertindole. E 4031 126-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-38 20097192-6 2010 KB130015 presumably binds to the hERG1 pore from the cytosolic side and functionally competes with hERG1 block by amiodarone, E4031 (N-[4-[[1-[2-(6-methyl-2-pyridinyl)ethyl]-4-piperidinyl] carbonyl] phenyl] methanesulfonamide dihydrochloride), and sertindole. E 4031 126-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-104 20097192-7 2010 Vice versa, amiodarone attenuates hERG1 activation by KB130015. Amiodarone 12-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-39 20097192-8 2010 Based on synergic channel activation by mallotoxin and KB130015 we conclude that the hERG1 pore contains at least two sites for activators that are functionally coupled among each other and to the cavity-blocker site. rottlerin 40-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-90 20097192-9 2010 KB130015 and amiodarone may serve as lead structures for the identification of hERG1 pore-interacting drugs favoring channel activation vs. block. Amiodarone 13-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-84 20015154-6 2010 Enrichment with cholesterol inhibited HERG currents, while inclusion of PIP2 in the pipette solution blocked the cholesterol effect. Cholesterol 16-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 19817928-2 2010 Vanoxerine is a drug that is free of adverse cardiac events in normal volunteers, yet is a potent blocker of the hERG (hK(v)11.1) cardiac potassium channel. vanoxerine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 119-128 19817928-6 2010 RESULTS: We found that vanoxerine was a potent hK(v)11.1 blocker, and at submicromolar concentrations, it blocked Ca and Na currents in a strongly frequency-dependent manner. vanoxerine 23-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-56 20390067-4 2010 Loss-of-function mutations in KCNH2, the gene encoding the cardiac ion channel that is responsible for the rapidly activating delayed rectifying potassium current, are linked to long-QT syndrome type 2 (LQT-2). Potassium 145-154 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-35 20390067-4 2010 Loss-of-function mutations in KCNH2, the gene encoding the cardiac ion channel that is responsible for the rapidly activating delayed rectifying potassium current, are linked to long-QT syndrome type 2 (LQT-2). Potassium 145-154 potassium voltage-gated channel subfamily H member 2 Homo sapiens 203-208 19765650-5 2010 In these cells blockade of HERG channels with the HERG blockers E 4301 or cisapride attenuated both proliferation and migration of the cells. Cisapride 74-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 19859662-0 2010 Regulation of HERG (KCNH2) potassium channel surface expression by diacylglycerol. Diglycerides 67-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 19859662-0 2010 Regulation of HERG (KCNH2) potassium channel surface expression by diacylglycerol. Diglycerides 67-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-25 19859662-3 2010 In the present report, using human embryonic kidney cells stably expressing HERG, we show that diacylglycerol potently inhibits the HERG current. Diglycerides 95-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 19859662-3 2010 In the present report, using human embryonic kidney cells stably expressing HERG, we show that diacylglycerol potently inhibits the HERG current. Diglycerides 95-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-136 19859662-6 2010 Thus, diacylglycerol is an important lipid participating in the regulation of HERG surface expression and function. Diglycerides 6-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 21063089-0 2010 The novel mechanism of oxymatrine affecting HERG currents at different temperatures. oxymatrine 23-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 19765650-5 2010 In these cells blockade of HERG channels with the HERG blockers E 4301 or cisapride attenuated both proliferation and migration of the cells. Cisapride 74-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 19765650-6 2010 Activation of HERG with PD118057 stimulated cell migration. 2-(4-(2-(3,4-dichlorophenyl)ethyl)phenylamino)benzoic acid 24-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 20210727-7 2010 Methadone is the opiod most tightly associated with QTc prolongation; with much lesser potency buprenorphine and oxycodone can block HERG channels and depress the IKr current in vitro.Antineoplastic chemotherapy like anthracyclines, alkylating drugs, alkilants and cisplatin are associated with electrocardiographic alterations including prolongation of QT and emesis of different grades. Buprenorphine 95-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 20210727-7 2010 Methadone is the opiod most tightly associated with QTc prolongation; with much lesser potency buprenorphine and oxycodone can block HERG channels and depress the IKr current in vitro.Antineoplastic chemotherapy like anthracyclines, alkylating drugs, alkilants and cisplatin are associated with electrocardiographic alterations including prolongation of QT and emesis of different grades. Oxycodone 113-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 19949665-3 2009 Application of MeOH extract of Lindera erythrocarpa showed a dose-dependent decrease in the amplitudes of the outward currents measured at the end of the pulse (I(HERG)) and the tail currents of HERG (I(tail)). Methanol 15-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 163-167 19577877-9 2010 The selective HERG1 inhibitor E-4031 dose-dependently impaired tumor growth in the proliferation assays. E 4031 30-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-19 19949665-3 2009 Application of MeOH extract of Lindera erythrocarpa showed a dose-dependent decrease in the amplitudes of the outward currents measured at the end of the pulse (I(HERG)) and the tail currents of HERG (I(tail)). Methanol 15-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 195-199 19949665-4 2009 When the BuOH fraction and H(2)O fraction of Lindera erythrocarpa were added to the perfusate, both I(HERG) and I(tail) were suppressed, while the hexane fraction, CHCl(3) fraction, and EtOAc fraction did not inhibit either I(HERG) or I(tail). Butanols 9-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 19949665-4 2009 When the BuOH fraction and H(2)O fraction of Lindera erythrocarpa were added to the perfusate, both I(HERG) and I(tail) were suppressed, while the hexane fraction, CHCl(3) fraction, and EtOAc fraction did not inhibit either I(HERG) or I(tail). Butanols 9-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 226-230 19892732-0 2009 PD-118057 contacts the pore helix of hERG1 channels to attenuate inactivation and enhance K+ conductance. 2-(4-(2-(3,4-dichlorophenyl)ethyl)phenylamino)benzoic acid 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-42 19671703-3 2009 Here we explored cyclic nucleotide binding and modulation of mEAG1 and hERG1 channels with fluorescence and electrophysiology. Nucleotides, Cyclic 17-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-76 19671703-4 2009 Binding of cyclic nucleotides to the isolated CNBD of mEAG1 and hERG1 channels was examined with two independent fluorescence-based methods: changes in tryptophan fluorescence and fluorescence of an analog of cAMP, 8-NBD-cAMP. Nucleotides, Cyclic 11-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-69 19671703-6 2009 Our results indicated that cyclic nucleotides do not bind to the isolated CNBD domain of mEAG1 channels and bind with low affinity (K(d) > or = 51 microm) to the isolated CNBD of hERG1 channels. Nucleotides, Cyclic 27-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 182-187 19725081-0 2009 Exploring chemical substructures essential for HERG k(+) channel blockade by synthesis and biological evaluation of dofetilide analogues. dofetilide 116-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 19457646-1 2009 Sertindole"s propensity to prolong the QT interval relates to blockade of the KCNH2 (HERG) encoded Ikr potassium channel, but there has been limited detailed data on T-wave morphology changes. sertindole 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-83 19457646-1 2009 Sertindole"s propensity to prolong the QT interval relates to blockade of the KCNH2 (HERG) encoded Ikr potassium channel, but there has been limited detailed data on T-wave morphology changes. sertindole 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 19892732-10 2009 Molecular modeling indicates that PD-118057 binds to a hydrophobic pocket formed by L646 of one hERG1 subunit and F619 of an adjacent subunit. 2-(4-(2-(3,4-dichlorophenyl)ethyl)phenylamino)benzoic acid 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-101 19892732-10 2009 Molecular modeling indicates that PD-118057 binds to a hydrophobic pocket formed by L646 of one hERG1 subunit and F619 of an adjacent subunit. 4-azaindole 84-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-101 19892732-11 2009 We conclude that direct interaction of PD-118057 with the pore helix attenuates fast P-type inactivation and increases open probability of hERG1 channels. 2-(4-(2-(3,4-dichlorophenyl)ethyl)phenylamino)benzoic acid 39-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-144 18692916-1 2009 In a 34-year-old man showing short QT interval (QTc 329 ms), we identified a novel C-terminal KCNH2 mutation, R1135H. qtc 48-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-99 19215240-6 2009 A mutant F463L HERG channel was expressed in HEK293 cells using a lipofectamine method. Lipofectamine 66-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19409379-4 2009 Our results showed that E-4031, a specific hERG1 K(+) channels inhibitor, significantly blocked SDF-1-induced migration of leukemic cell lines, primary acute leukemic cells, leukemic stem cells and HEK293T cells transfected with herg-pEGFP. E 4031 24-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-48 19424681-0 2009 Blockade of HERG K+ channel by isoquinoline alkaloid neferine in the stable transfected HEK293 cells. Tubocurarine 31-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 19424681-0 2009 Blockade of HERG K+ channel by isoquinoline alkaloid neferine in the stable transfected HEK293 cells. neferine 53-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 19424681-1 2009 We studied the effects of isoquinoline alkaloid neferine (Nef) extracted from the seed embryo of Nelumbo nucifera Gaertn on Human ether-a-go-go-related gene (HERG) channels stably expressed in human embryonic kidney (HEK293) cells using whole-cell patch clamp technique, western blot analysis and immunofluorescence experiment. Tubocurarine 26-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 158-162 19424681-1 2009 We studied the effects of isoquinoline alkaloid neferine (Nef) extracted from the seed embryo of Nelumbo nucifera Gaertn on Human ether-a-go-go-related gene (HERG) channels stably expressed in human embryonic kidney (HEK293) cells using whole-cell patch clamp technique, western blot analysis and immunofluorescence experiment. neferine 48-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 158-162 19419905-3 2009 OBJECTIVE: We investigated acute alpha(1A)- and cyclic adenosine monophosphate (cAMP)-related beta-adrenergic modulation of I(Kr) in HL-1 cardiomyocytes, wild type (WT)- and 2 LQT2-associated mutant Kv11.1 channels (Y43D- and K595E-Kv11.1) reconstituted in Chinese hamster ovary (CHO) cells. Cyclic AMP 80-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 176-180 19617705-0 2009 Extracellular potassium dependency of block of HERG by quinidine and cisapride is primarily determined by the permeant ion and not by inactivation. Potassium 14-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 19617705-0 2009 Extracellular potassium dependency of block of HERG by quinidine and cisapride is primarily determined by the permeant ion and not by inactivation. Quinidine 55-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 19617705-0 2009 Extracellular potassium dependency of block of HERG by quinidine and cisapride is primarily determined by the permeant ion and not by inactivation. Cisapride 69-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. Quinidine 29-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. Cisapride 43-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. Calcium 96-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. Potassium 105-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. Rubidium 116-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. cesium and tetraethylammonium 126-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-6 2009 In this study, HERG block by quinidine and cisapride was assessed in extracellular solutions of calcium, potassium, rubidium, cesium and tetraethylammonium (TEA) using two-electrode voltage clamping of Xenopus oocytes. Tetraethylammonium 157-160 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 19617705-7 2009 Consistent with previous reports we show that increases in extracellular potassium reduce HERG block by quinidine and cisapride. Potassium 73-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 19617705-7 2009 Consistent with previous reports we show that increases in extracellular potassium reduce HERG block by quinidine and cisapride. Quinidine 104-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 19617705-7 2009 Consistent with previous reports we show that increases in extracellular potassium reduce HERG block by quinidine and cisapride. Cisapride 118-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 19617705-8 2009 We also show that increasing extracellular rubidium and cesium reduced HERG block by quinidine and cisapride whereas increasing extracellular calcium and extracellular TEA did not alter HERG block by quinidine and cisapride. Rubidium 43-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 19617705-8 2009 We also show that increasing extracellular rubidium and cesium reduced HERG block by quinidine and cisapride whereas increasing extracellular calcium and extracellular TEA did not alter HERG block by quinidine and cisapride. Cesium 56-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 19617705-8 2009 We also show that increasing extracellular rubidium and cesium reduced HERG block by quinidine and cisapride whereas increasing extracellular calcium and extracellular TEA did not alter HERG block by quinidine and cisapride. Quinidine 85-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 19617705-8 2009 We also show that increasing extracellular rubidium and cesium reduced HERG block by quinidine and cisapride whereas increasing extracellular calcium and extracellular TEA did not alter HERG block by quinidine and cisapride. Cisapride 99-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 19617705-9 2009 These results demonstrate that at lower extracellular potassium concentrations, the permeant ion is almost exclusively responsible for the reduction in quinidine and cisapride block of HERG due to increases in extracellular potassium. Potassium 54-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 19617705-9 2009 These results demonstrate that at lower extracellular potassium concentrations, the permeant ion is almost exclusively responsible for the reduction in quinidine and cisapride block of HERG due to increases in extracellular potassium. Cisapride 166-175 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 19617705-9 2009 These results demonstrate that at lower extracellular potassium concentrations, the permeant ion is almost exclusively responsible for the reduction in quinidine and cisapride block of HERG due to increases in extracellular potassium. Potassium 224-233 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 19528813-6 2009 The IC50 for quinidine block of IKr in CHO cells transfected with HERG alone was significantly higher than cells transfected with HERG + OCTN1 (0.66 +/- 0.15 microM versus 0.14 +/- 0.06 microM [52% absolute increase in drug block; 95% confidence interval, 0.4-0.64 microM]), and this effect was further potentiated by a common genetic variant of OCTN1, L503F. Quinidine 13-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 19366693-3 2009 One of the identified compounds, gambogic acid (GA), potently (EC(50) < or = 100 nm) inhibited Kir2.1 channels in mammalian cells when applied chronically for 3 h. This potent and slow inhibition was not seen with Kv2.1, HERG or Kir1.1 channels. gambogic acid 33-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 224-228 19366693-3 2009 One of the identified compounds, gambogic acid (GA), potently (EC(50) < or = 100 nm) inhibited Kir2.1 channels in mammalian cells when applied chronically for 3 h. This potent and slow inhibition was not seen with Kv2.1, HERG or Kir1.1 channels. gambogic acid 48-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 224-228 18998156-6 2009 Gating of erg1 channels kept the voltage response to glutamate brief and at physiological amplitudes. Glutamic Acid 53-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 10-14 19406877-0 2009 The evolutionarily conserved residue A653 plays a key role in HERG channel closing. a653 37-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 19406877-3 2009 Here, we assess the functional role of the alanine at position 653 (HERG-A653) that is highly conserved among evolutionarily divergent K(+) channels. Alanine 43-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 19406877-4 2009 HERG-A653 is close to the "glycine hinge" implicated in K(+) channel opening, and is flanked by tyrosine 652 and phenylalanine 656, which contribute to the drug binding site. Glycine 27-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 19406877-4 2009 HERG-A653 is close to the "glycine hinge" implicated in K(+) channel opening, and is flanked by tyrosine 652 and phenylalanine 656, which contribute to the drug binding site. Tyrosine 96-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 19406877-4 2009 HERG-A653 is close to the "glycine hinge" implicated in K(+) channel opening, and is flanked by tyrosine 652 and phenylalanine 656, which contribute to the drug binding site. Phenylalanine 113-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 19406877-10 2009 Our study suggests that A653 plays an important functional role in the outwardly rectifying gating behaviour of HERG, supporting channel closure at membrane potentials negative to the channel activation threshold. a653 24-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-116 19204737-0 2009 Influence of the G2677T/C3435T haplotype of MDR1 on P-glycoprotein trafficking and ibutilide-induced block of HERG. ibutilide 83-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 19204737-3 2009 We tested the effect of these MDR1 variants on Human Ether-Related A Go-Go (HERG) block by ibutilide in CHO cells 48 h following transient transfection with an IRES-dsRed vector containing MDR1, G2677T MDR1, G2677T/C3435T MDR1 or an empty bicistronic site and an IRES-GFP vector containing HERG (KCNH2). ibutilide 91-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 19204737-3 2009 We tested the effect of these MDR1 variants on Human Ether-Related A Go-Go (HERG) block by ibutilide in CHO cells 48 h following transient transfection with an IRES-dsRed vector containing MDR1, G2677T MDR1, G2677T/C3435T MDR1 or an empty bicistronic site and an IRES-GFP vector containing HERG (KCNH2). ibutilide 91-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19204737-3 2009 We tested the effect of these MDR1 variants on Human Ether-Related A Go-Go (HERG) block by ibutilide in CHO cells 48 h following transient transfection with an IRES-dsRed vector containing MDR1, G2677T MDR1, G2677T/C3435T MDR1 or an empty bicistronic site and an IRES-GFP vector containing HERG (KCNH2). CAV protocol 104-107 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 19204737-8 2009 Coculture with fexofenadine(1 microM), an MDR1 substrate known to rescue misfolding in other membrane proteins, restored cell surface expression of MDR1 G2677T/C3435T and restored resistance to block HERG by ibutilide 200 nM (98.5+/-0.98% vs 42.3+/-2.2%, P<0.001). fexofenadine 15-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-204 19204737-8 2009 Coculture with fexofenadine(1 microM), an MDR1 substrate known to rescue misfolding in other membrane proteins, restored cell surface expression of MDR1 G2677T/C3435T and restored resistance to block HERG by ibutilide 200 nM (98.5+/-0.98% vs 42.3+/-2.2%, P<0.001). ibutilide 208-217 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-204 19204737-10 2009 These data identify ibutilide as an MDR1 substrate and further support the concept that variable drug transport function can modulate the action of HERG blockers. ibutilide 20-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 19413965-1 2009 hERG1 is a member of the cyclic nucleotide binding domain family of K(+) channels. Nucleotides, Cyclic 25-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-5 19413965-3 2009 We reasoned that histidine 562 in hERG1 could play an important structure-function role. Histidine 17-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-39 19413965-4 2009 To explore this role, we created in silica models of the hERG1 pore domain based on the KvAP crystal structure with Rosetta-membrane modeling and molecular-dynamics simulations. Silicon Dioxide 36-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-62 19413965-11 2009 In conclusion, interactions between H562 in the S5 helix and amino acids in the pore helix are important determinants of hERG1 potassium channel function, as confirmed by theory and experiment. 7-methylquinazoline-2,4-diamine 36-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-126 19234087-6 2009 Direct PKA phosphorylation of the HERG protein was responsible for the cAMP-induced augmentation. Cyclic AMP 71-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 19413965-0 2009 Interactions of H562 in the S5 helix with T618 and S621 in the pore helix are important determinants of hERG1 potassium channel structure and function. 7-methylquinazoline-2,4-diamine 16-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-109 19234087-9 2009 Acceleration of the HERG protein synthesis rate was the primary factor in the cAMP/PKA effect with lesser effects on protein stability. Cyclic AMP 78-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 19244476-0 2009 State-dependent block of HERG potassium channels by R-roscovitine: implications for cancer therapy. Roscovitine 52-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 19285067-7 2009 Furthermore, PF-01354082 decreased I(HERG) current by only 11% at a concentration of 300 microM, indicating that the compound had greater than 150,000-fold selectivity for the human 5-HT(4d) receptor over hERG channels. 4-((4-((((3-Isopropyl-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl)carbonyl)amino)methyl)piperidin-1-yl)methyl)tetrahydro-2H-pyran-4-carboxylic acid 13-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 19244476-6 2009 In the present study we show that R-roscovitine blocks HERG potassium current (human embryonic kidney-293 cells stably expressing HERG) at clinically relevant concentrations. Roscovitine 34-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 19244476-6 2009 In the present study we show that R-roscovitine blocks HERG potassium current (human embryonic kidney-293 cells stably expressing HERG) at clinically relevant concentrations. Roscovitine 34-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 19244476-6 2009 In the present study we show that R-roscovitine blocks HERG potassium current (human embryonic kidney-293 cells stably expressing HERG) at clinically relevant concentrations. Potassium 60-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 19244476-8 2009 Kinetic study of wild-type and inactivation mutant HERG channels supported block of activated channels by roscovitine with relatively little effect on either closed or inactivated channels. Roscovitine 106-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 19244476-9 2009 A HERG gating model reproduced all roscovitine effects. Roscovitine 35-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 2-6 19324319-0 2009 Aminoglycoside antibiotics restore functional expression of truncated HERG channels produced by nonsense mutations. Aminoglycosides 0-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Cyclic AMP 23-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Cyclic AMP 23-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). chlorophenylthiol 38-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). chlorophenylthiol 38-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). cpt 57-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). cpt 57-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Cyclic AMP 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Cyclic AMP 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Colforsin 70-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Colforsin 70-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Cyclic AMP 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 19234087-4 2009 Sustained elevation of cAMP by either chlorophenylthiol (CPT)-cAMP or forskolin increased the HERG channel protein abundance two- to fourfold within 24 h, with measurable difference as early as 4 h. The cAMP-induced augmentation was not due to changes in transcription and was specific for HERG compared with other cardiac K(+) channels (Kv1.4, Kv1.5, Kir2.1, and KvLQT1). Cyclic AMP 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 290-294 19324319-3 2009 OBJECTIVE: The purpose of this study was to investigate the effect of aminoglycoside antibiotics on the expression of nonsense mutants expected to produce truncated HERG channels. Aminoglycosides 70-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-169 19374880-0 2009 Pitavastatin suppresses mitogen activated protein kinase-mediated Erg-1 induction in human vascular smooth muscle cells. pitavastatin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-71 19038263-6 2009 Here we show that I(Kr) recorded in cells co-expressing HERG and ARHGAP6 was decreased by 43% compared to HERG alone. arhgap6 65-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 19038263-7 2009 Biochemical measurements of cell-surface associated HERG revealed that ARHGAP6 reduced membrane expression of HERG by 35%, which correlates well with the reduction in current. arhgap6 71-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 19038263-7 2009 Biochemical measurements of cell-surface associated HERG revealed that ARHGAP6 reduced membrane expression of HERG by 35%, which correlates well with the reduction in current. arhgap6 71-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 19038263-11 2009 We further determined that ARHGAP6 effects are mediated by a consensus SH3 binding domain within the C-terminus of HERG, although stable ARHGAP6-HERG complexes were not observed. arhgap6 27-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 19129680-0 2009 A receptor-independent effect of estrone sulfate on the HERG channel. estrone sulfate 33-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 18984955-7 2009 RESULTS: At concentrations of 0.01, 0.03, 0.1, 1 and 3 mM, r/s-methadone produced a dose-dependent inhibition of HERG by 17 +/- 5, 23 +/- 4, 40 +/- 4, 57 +/- 3, 69 +/- 3 and 80 +/- 1%, respectively. r/s-methadone 59-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 18984955-9 2009 At 0.1, 0.3 and 1 mM, s-methadone reduced HERG current by 50 +/- 4, 76 +/- 5 and 87 +/- 5%, respectively, while r-methadone reduced HERG current by 26 +/- 4, 53 +/- 3 and 77 +/- 3%, respectively. Methadone 22-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 18984955-9 2009 At 0.1, 0.3 and 1 mM, s-methadone reduced HERG current by 50 +/- 4, 76 +/- 5 and 87 +/- 5%, respectively, while r-methadone reduced HERG current by 26 +/- 4, 53 +/- 3 and 77 +/- 3%, respectively. Methadone 112-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-136 19530746-17 2009 In subjects receiving sotalol, T-wave morphology reached similarity to LQT2, whereas QTcF did not. Sotalol 22-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 19530746-18 2009 CONCLUSION: Distinct ECG patterns in LQT2 carriers effectively quantified repolarization changes induced by sotalol. Sotalol 108-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 18781376-2 2009 Here, we investigated the mechanism of hERG K(+) channel current (I(hERG)) blockade expressed in HEK-293 cells by sibutramine HCl, a serotonin-norepinephrine reuptake inhibitor. sibutramine 114-129 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-73 19143746-3 2009 To study whether acehytisine hydrochloride affects HERG channel activity and protein trafficking, we expressed HERG in human embryonic kidney 293 (HEK293) cells and recorded HERG channel currents with a whole-cell patch clamp technique. acehytisine hydrochloride 17-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 19143746-5 2009 We found that acehytisine hydrochloride inhibited HERG step current (I(HERG)) in a concentration-dependent manner. acehytisine hydrochloride 14-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 19143746-5 2009 We found that acehytisine hydrochloride inhibited HERG step current (I(HERG)) in a concentration-dependent manner. acehytisine hydrochloride 14-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 19143746-9 2009 Moreover, the F656C mutation in the S6 domain abolished acehytisine hydrochloride inhibition on the I(HERG) and enhanced the inhibitive effects on the trafficking of the 155 kD band. acehytisine hydrochloride 56-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 19143746-10 2009 Acehytisine hydrochloride interrupted HERG protein trafficking at 1000 and 2500 microM. acehytisine hydrochloride 0-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 19143746-11 2009 Our data showed that acehytisine hydrochloride could inhibit I(HERG), but had no effects on I(tail) until the concentration was above 1000 microM. acehytisine hydrochloride 21-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 19034039-0 2008 Papaverine, a vasodilator, blocks the pore of the HERG channel at submicromolar concentration. Papaverine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 19034039-2 2008 To examine the mechanism for this effect, we herein tested the effects of papaverine on human ether-a-go-go (HERG) K channels expressed in HEK293 cells and Xenopus oocytes. Papaverine 74-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 19034039-3 2008 Our results revealed that papaverine dose-dependently decreased the tail currents of HERG channel expressed in HEK293 cells with the IC50 and the Hill coefficient of 0.58 microM and 0.58, respectively, at +20 mV and 36 degrees C. The IC50 for the papaverine-induced blockade of HERG current in Xenopus oocytes was found to decrease progressively relative to depolarization (38.8, 30.0, and 24.8 microM at -10, +20, and +40 mV, respectively). Papaverine 26-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 19034039-3 2008 Our results revealed that papaverine dose-dependently decreased the tail currents of HERG channel expressed in HEK293 cells with the IC50 and the Hill coefficient of 0.58 microM and 0.58, respectively, at +20 mV and 36 degrees C. The IC50 for the papaverine-induced blockade of HERG current in Xenopus oocytes was found to decrease progressively relative to depolarization (38.8, 30.0, and 24.8 microM at -10, +20, and +40 mV, respectively). Papaverine 26-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 278-282 19034039-3 2008 Our results revealed that papaverine dose-dependently decreased the tail currents of HERG channel expressed in HEK293 cells with the IC50 and the Hill coefficient of 0.58 microM and 0.58, respectively, at +20 mV and 36 degrees C. The IC50 for the papaverine-induced blockade of HERG current in Xenopus oocytes was found to decrease progressively relative to depolarization (38.8, 30.0, and 24.8 microM at -10, +20, and +40 mV, respectively). Papaverine 247-257 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 19034039-4 2008 The papaverine-induced blockade of HERG current was time-dependent; the fractional current was 0.92 +/- 0.03 of the control at the beginning of the pulse, but it declined to 0.18 +/- 0.06 after 6 seconds at a test potential of 0 mV. Papaverine 4-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 19034039-5 2008 These results collectively indicate that papaverine blocks HERG channel in a concentration-, voltage-, and time-dependent manner. Papaverine 41-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 19034039-6 2008 Two S6 domain mutations, Y652A and F656A, partially attenuated (Y652A) or abolished (F656A) the hERG current blockade, suggesting that papaverine blocks HERG channel at the pore of the channel. Papaverine 135-145 potassium voltage-gated channel subfamily H member 2 Homo sapiens 153-157 19034039-8 2008 Therefore, ventricular arrhythmias induced by papaverine could be resulted from the blockage of the HERG channel at the cardiac myocytes. Papaverine 46-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 19057127-0 2008 Hydroxyzine, a first generation H(1)-receptor antagonist, inhibits human ether-a-go-go-related gene (HERG) current and causes syncope in a patient with the HERG mutation. Hydroxyzine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 19057127-0 2008 Hydroxyzine, a first generation H(1)-receptor antagonist, inhibits human ether-a-go-go-related gene (HERG) current and causes syncope in a patient with the HERG mutation. Hydroxyzine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 156-160 19057127-2 2008 In this paper, we will report a case of drug-induced long QT syndrome associated with an H(1)-receptor antagonist, hydroxyzine, in which a mutation was identified in the HERG gene. Hydroxyzine 115-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 170-174 19057127-7 2008 Hydroxyzine concentration-dependently inhibited both wild-type (WT) and WT/A614V-HERG K(+) currents. Hydroxyzine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 18781376-2 2009 Here, we investigated the mechanism of hERG K(+) channel current (I(hERG)) blockade expressed in HEK-293 cells by sibutramine HCl, a serotonin-norepinephrine reuptake inhibitor. Serotonin 133-142 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-73 18781376-2 2009 Here, we investigated the mechanism of hERG K(+) channel current (I(hERG)) blockade expressed in HEK-293 cells by sibutramine HCl, a serotonin-norepinephrine reuptake inhibitor. Norepinephrine 143-157 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-73 18805669-7 2008 Berberine could reduce SDF-1 protein level secreted by BMSCs in the microenvironment but not affect CXCR4 expression on HL-60 cell membrane, and we hypothesized that berberine could inhibit AML cells migration partly by reducing the secreting of SDF-1 by BMSCs and inhibiting HERG1 K(+) channels of leukemic cells. Berberine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 276-281 18724381-12 2008 In addition, our findings indicate that amiodarone may warrant further investigation as a potential treatment for SQT1. Amiodarone 40-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 18805669-7 2008 Berberine could reduce SDF-1 protein level secreted by BMSCs in the microenvironment but not affect CXCR4 expression on HL-60 cell membrane, and we hypothesized that berberine could inhibit AML cells migration partly by reducing the secreting of SDF-1 by BMSCs and inhibiting HERG1 K(+) channels of leukemic cells. Berberine 166-175 potassium voltage-gated channel subfamily H member 2 Homo sapiens 276-281 18599551-3 2008 In a concentration-dependent manner (10(-9) to 10(-6) M for 24 h), 5alpha-dihydrotestosterone (5alpha-DHT) increased HERG protein abundance in HEK293 cells stably expressing HERG in the presence of coexpressed cardiac androgen receptor (AR) variant [N-terminal truncated isoform of AR (AR45)]. Dihydrotestosterone 67-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 18599551-3 2008 In a concentration-dependent manner (10(-9) to 10(-6) M for 24 h), 5alpha-dihydrotestosterone (5alpha-DHT) increased HERG protein abundance in HEK293 cells stably expressing HERG in the presence of coexpressed cardiac androgen receptor (AR) variant [N-terminal truncated isoform of AR (AR45)]. Dihydrotestosterone 67-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 18599551-3 2008 In a concentration-dependent manner (10(-9) to 10(-6) M for 24 h), 5alpha-dihydrotestosterone (5alpha-DHT) increased HERG protein abundance in HEK293 cells stably expressing HERG in the presence of coexpressed cardiac androgen receptor (AR) variant [N-terminal truncated isoform of AR (AR45)]. Dihydrotestosterone 95-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 18599551-3 2008 In a concentration-dependent manner (10(-9) to 10(-6) M for 24 h), 5alpha-dihydrotestosterone (5alpha-DHT) increased HERG protein abundance in HEK293 cells stably expressing HERG in the presence of coexpressed cardiac androgen receptor (AR) variant [N-terminal truncated isoform of AR (AR45)]. Dihydrotestosterone 95-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 18599551-6 2008 Proteasome inhibitors, N-acetyl-L-leucyl-L-leucyl-L-norleucinal and MG132 prevented the 5alpha-DHT- dependent enhancement of HERG, as did the lysosome inhibitor, bafilomycin A1. Cysteine 25-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 18599551-6 2008 Proteasome inhibitors, N-acetyl-L-leucyl-L-leucyl-L-norleucinal and MG132 prevented the 5alpha-DHT- dependent enhancement of HERG, as did the lysosome inhibitor, bafilomycin A1. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 68-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 18599551-6 2008 Proteasome inhibitors, N-acetyl-L-leucyl-L-leucyl-L-norleucinal and MG132 prevented the 5alpha-DHT- dependent enhancement of HERG, as did the lysosome inhibitor, bafilomycin A1. Dihydrotestosterone 95-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 18599551-7 2008 Consistently, the cycloheximide-based protein chase study showed that 5alpha-DHT prolonged HERG protein half-life. Cycloheximide 18-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 18599551-7 2008 Consistently, the cycloheximide-based protein chase study showed that 5alpha-DHT prolonged HERG protein half-life. Dihydrotestosterone 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 18599551-9 2008 Blockade of ERK1/2 with PD98059 and U0126 prevented the effect of androgen on HERG protein abundance. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 24-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 18599551-9 2008 Blockade of ERK1/2 with PD98059 and U0126 prevented the effect of androgen on HERG protein abundance. U 0126 36-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 18599551-10 2008 Functional studies showed that 5alpha-DHT treatment for 24 h increased HERG K+ current density in Chinese hamster ovary cells cotransfected with cDNAs of AR45 and HERG channels. Dihydrotestosterone 31-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 18599551-10 2008 Functional studies showed that 5alpha-DHT treatment for 24 h increased HERG K+ current density in Chinese hamster ovary cells cotransfected with cDNAs of AR45 and HERG channels. Dihydrotestosterone 31-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 163-167 19024448-9 2008 Thus, altough the kinetics properties of HERG channels were complicated, the channels kinetics could be indirectly analyzed through differently designed pulse protocols, which provided the basis for investigation on Alanine-scanning mutagenesis and agent action. Alanine 216-223 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 18791070-3 2008 Here, we show that the hERG1 polymorphism at codon 897, which is read as a Thr instead of a Lys, creates a phosphorylation site for the Akt protein kinase on the Kv11.1 channel protein. Threonine 75-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-28 18808722-9 2008 KCNH2 SNP K897T was reported to exert a modifying effect on QTc, but it remains controversial whether it confers a risk or protective effect. qtc 60-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-5 18713925-1 2008 Inhibition of the HERG channel by droperidol depends on channel gating and involves the S6 residue F656. Droperidol 34-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 18713925-1 2008 Inhibition of the HERG channel by droperidol depends on channel gating and involves the S6 residue F656. f656 99-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 18791070-3 2008 Here, we show that the hERG1 polymorphism at codon 897, which is read as a Thr instead of a Lys, creates a phosphorylation site for the Akt protein kinase on the Kv11.1 channel protein. Threonine 75-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-168 18791070-3 2008 Here, we show that the hERG1 polymorphism at codon 897, which is read as a Thr instead of a Lys, creates a phosphorylation site for the Akt protein kinase on the Kv11.1 channel protein. Lysine 92-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-28 18791070-3 2008 Here, we show that the hERG1 polymorphism at codon 897, which is read as a Thr instead of a Lys, creates a phosphorylation site for the Akt protein kinase on the Kv11.1 channel protein. Lysine 92-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-168 18664324-0 2008 Electropharmacological properties of telmisartan in blocking hKv1.5 and HERG potassium channels expressed on Xenopus laevis oocytes. Telmisartan 37-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 18690396-1 2008 In the present study, we investigated the inhibitory action of ketanserin on wild-type (WT) and Y652 mutant human ether-a-go-go-related gene (HERG) potassium channels expressed in Xenopus oocytes and the effects of changing the channel molecular determinants characteristics on the blockade with and without ketanserin intervention using standard two-microelectrode voltage-clamp techniques. Ketanserin 63-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 18690396-5 2008 The results showed that ketanserin blocked WT HERG currents in voltage- and concentration-dependent manner and showed minimal tonic blockade of HERG current evaluated by the envelope of tails test. Ketanserin 24-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 18690396-10 2008 Ketanserin could also modulate the inactivation of HERG channel, which shifted the voltage-dependence of WT HERG channel inactivation curve from (-51.71+/-2.15) mV to (-80.76+/-14.98) mV (P<0.05, n=4). Ketanserin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 18690396-14 2008 In conclusion, ketanserin blocks WT HERG currents in voltage- and concentration-dependent manner and preferentially blocks open-state HERG channels. Ketanserin 15-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 18690396-14 2008 In conclusion, ketanserin blocks WT HERG currents in voltage- and concentration-dependent manner and preferentially blocks open-state HERG channels. Ketanserin 15-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 134-138 18664324-1 2008 AIM: The objectives of this study were to investigate the inhibitory action of telmisartan, a selective angiotensin II type 1 receptor antagonist, on hKv1.5 and human ether-a-go-go-related gene (HERG) channels expressed on Xenopus laevis oocytes. Telmisartan 79-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 195-199 18664324-5 2008 However, telmisartan exhibited a low affinity for HERG channels (IC50=24.35+/-5.06 micromol/L); the blockade was voltage- and concentration-dependent. Telmisartan 9-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 18458880-10 2008 HERG inactivation kinetics were significantly altered by mianserin without marked effects on channel activation kinetics. Mianserin 57-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 18349188-0 2008 Inhibition of the HERG channel by droperidol depends on channel gating and involves the S6 residue F656. Droperidol 34-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 18520952-0 2008 Rapid kinetic interactions of ranolazine with HERG K+ current. Ranolazine 30-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 18520952-1 2008 Ranolazine, an anti-ischemic agent, inhibits I Kr [encoded by the human ether-a-go-go-related gene (HERG)] and causes a small QT interval prolongation without any proarrhythmic events. Ranolazine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 18520952-2 2008 The objective of this study was to elucidate the biophysical characteristics of inhibition of HERG K+ current (IHERG) by ranolazine. Ranolazine 121-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 18690396-10 2008 Ketanserin could also modulate the inactivation of HERG channel, which shifted the voltage-dependence of WT HERG channel inactivation curve from (-51.71+/-2.15) mV to (-80.76+/-14.98) mV (P<0.05, n=4). Ketanserin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 18679741-3 2008 The HERG potassium channel, which produces the cardiac rapidly activating delayed rectifying K+ current (I(Kr)), is a target for cAMP/PKA regulation. Cyclic AMP 129-133 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 18679741-7 2008 The functional consequence of AKAP-IS is a reversal of cAMP-dependent regulation of HERG channel activity. Cyclic AMP 55-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 18679741-11 2008 These results suggest that one or more AKAP(s) targets PKA to HERG channels and may contribute to the acute regulation of I(Kr) by cAMP. Cyclic AMP 131-135 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 18379053-0 2008 HERG K+ channel blockade by the novel antiviral drug sophocarpine. sophocarpine 53-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 18379053-3 2008 The effects of the novel antiviral drug sophocarpine (SC) were examined on stably expressed HERG channels in human embryonic kidney (HEK293) cells using a whole-cell patch clamp technique, Western blot analysis and immunofluorescence experiments. sophocarpine 40-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 18379053-3 2008 The effects of the novel antiviral drug sophocarpine (SC) were examined on stably expressed HERG channels in human embryonic kidney (HEK293) cells using a whole-cell patch clamp technique, Western blot analysis and immunofluorescence experiments. sophocarpine 54-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 18349188-0 2008 Inhibition of the HERG channel by droperidol depends on channel gating and involves the S6 residue F656. f656 99-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 18349188-5 2008 To determine the mechanism underlying these clinical findings, we investigated the effect of droperidol on human HERG potassium channels. Droperidol 93-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 18349188-7 2008 RESULTS: HERG tail currents following test pulses to 50 mV were inhibited by droperidol with an IC(50) of 77.3 +/- 9.6 nM (n = 8). Droperidol 77-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 18349188-9 2008 Droperidol affected HERG channels mainly in their open and inactivated states. Droperidol 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 18349188-13 2008 The potency for block of HERG channels by droperidol was significantly decreased with mutation of Phe-656 to Thr or mutation of Ser-631 to Ala, respectively. Droperidol 42-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 18349188-15 2008 CONCLUSIONS: Droperidol potently inhibits transfected HERG channels and this is the probable mechanism for QT prolongation. Droperidol 13-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 18006430-10 2008 Amiodarone inhibited HERG K+ and late Na+ currents with IC50s of 0.8 +/- 0.1 and 3.0 +/- 0.9 microM, respectively. Amiodarone 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 17516459-0 2008 Blockade of HERG K+ channel by an antihistamine drug brompheniramine requires the channel binding within the S6 residue Y652 and F656. Brompheniramine 53-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 17516459-2 2008 This study investigated the molecular mechanisms of voltage-dependent inhibition of human ether-a-go-go-related gene (HERG) delayed rectifier K+ channels expressed in HEK-293 cells by brompheniramine, an antihistamine. Brompheniramine 184-199 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-122 17516459-3 2008 Brompheniramine inhibited HERG current in a concentration-dependent manner with the half-maximal inhibitory concentration (IC50) value of 1.7 microm at 0 mV. Brompheniramine 0-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 17516459-4 2008 A block of HERG current by brompheniramine was enhanced by progressive membrane depolarization and showed significantly negative shift in voltage-dependence of channel activation. Brompheniramine 27-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 11-15 17516459-5 2008 Inhibition of HERG current by brompheniramine showed time-dependence. Brompheniramine 30-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 17516459-7 2008 These results indicate that brompheniramine mainly inhibited the HERG potassium channel through the residue Y652 and F656 and these residues may be an obligatory determinant in inhibition of HERG current for brompheniramine. Brompheniramine 28-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 17516459-7 2008 These results indicate that brompheniramine mainly inhibited the HERG potassium channel through the residue Y652 and F656 and these residues may be an obligatory determinant in inhibition of HERG current for brompheniramine. Brompheniramine 28-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 191-195 17516459-7 2008 These results indicate that brompheniramine mainly inhibited the HERG potassium channel through the residue Y652 and F656 and these residues may be an obligatory determinant in inhibition of HERG current for brompheniramine. Brompheniramine 208-223 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 17516459-7 2008 These results indicate that brompheniramine mainly inhibited the HERG potassium channel through the residue Y652 and F656 and these residues may be an obligatory determinant in inhibition of HERG current for brompheniramine. Brompheniramine 208-223 potassium voltage-gated channel subfamily H member 2 Homo sapiens 191-195 18181608-0 2008 Incorporation of the HERG potassium channel in a mercury supported lipid bilayer. Mercury 49-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 18191158-0 2008 Protriptyline block of the human ether-a-go-go-related gene (HERG) K+ channel. Protriptyline 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-65 18191158-2 2008 We studied the effects of protriptyline on human ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes and HEK293 cells. Protriptyline 26-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 18191158-3 2008 Protriptyline induced a concentration-dependent decrease in current amplitudes at the end of the voltage steps and HERG tail currents. Protriptyline 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 18092154-7 2008 Propiverine blocked in a concentration-dependent manner HERG channels expressed in HEK293 cells, as well as native I(Kr) current in ventricular myocytes of guinea pig (IC50 values: 10 microM and 1.8 microM respectively). propiverine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 18181608-1 2008 The HERG potassium channel was incorporated in a mercury-supported tethered bilayer lipid membrane (tBLM) obtained by anchoring a thiolipid monolayer to the mercury surface and by self-assembling a lipid monolayer on top of it from a lipid film spread on the surface of an electrolyte solution. Mercury 49-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 18181608-1 2008 The HERG potassium channel was incorporated in a mercury-supported tethered bilayer lipid membrane (tBLM) obtained by anchoring a thiolipid monolayer to the mercury surface and by self-assembling a lipid monolayer on top of it from a lipid film spread on the surface of an electrolyte solution. tblm 100-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 18191158-4 2008 The IC(50) for protriptyline block of HERG current in Xenopus oocytes progressively decreased relative to the degree of depolarization, from 142.0 microM at -40 mV to 91.7 microM at 0 mV to 52.9 microM at +40 mV. Protriptyline 15-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 18191158-6 2008 The IC(50) for the protriptyline-induced blockade of HERG currents in HEK293 cells at 36 degrees C was 1.18 microM at +20 mV. Protriptyline 19-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 53-57 18191158-8 2008 HERG blockade by protriptyline was use-dependent, exhibiting a more rapid onset and a greater steady-state block at higher frequencies of activation. Protriptyline 17-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 18191158-9 2008 Our findings suggest that inhibition of HERG currents may contribute to the arrhythmogenic side effects of protriptyline. Protriptyline 107-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 18057887-0 2008 Extracellular acidification and hyperkalemia induce changes in HERG inhibition by ibutilide. ibutilide 82-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 18181608-1 2008 The HERG potassium channel was incorporated in a mercury-supported tethered bilayer lipid membrane (tBLM) obtained by anchoring a thiolipid monolayer to the mercury surface and by self-assembling a lipid monolayer on top of it from a lipid film spread on the surface of an electrolyte solution. thiolipid 130-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 18057887-2 2008 Our previous studies have shown that extracellular acidosis and hyperkalemia attenuate the HERG-inhibitory effect of proarrhythmic drugs, e.g. quinidine, but have little impact on the less-proarrhythmic drug amiodarone. Quinidine 143-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 18181608-1 2008 The HERG potassium channel was incorporated in a mercury-supported tethered bilayer lipid membrane (tBLM) obtained by anchoring a thiolipid monolayer to the mercury surface and by self-assembling a lipid monolayer on top of it from a lipid film spread on the surface of an electrolyte solution. Mercury 157-164 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 18181608-2 2008 HERG was then incorporated in this tBLM from its micellar solution in Triton X-100, thus avoiding the use of vesicles in the preparation of the tBLM and of proteoliposomes in channel incorporation. tblm 35-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 18181608-2 2008 HERG was then incorporated in this tBLM from its micellar solution in Triton X-100, thus avoiding the use of vesicles in the preparation of the tBLM and of proteoliposomes in channel incorporation. Octoxynol 70-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 18181608-2 2008 HERG was then incorporated in this tBLM from its micellar solution in Triton X-100, thus avoiding the use of vesicles in the preparation of the tBLM and of proteoliposomes in channel incorporation. tblm 144-148 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 19088442-0 2008 A highly conserved alanine in the S6 domain of the hERG1 K+ channel is required for normal gating. Alanine 19-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-56 19088442-3 2008 In hERG1 channels, we determined the functional consequences of substituting this conserved Ala (Ala653) with other hydrophobic or charged amino acids. ala 92-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-8 19088442-10 2008 Thus, an Ala at position 653 in hERG1 is required for normal voltage dependence of channel gating and a charged residue in this position prevents channel closure. ala 9-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-37 18357730-0 2008 [Effect of sophocarpine on HERG K+ channels]. sophocarpine 11-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 18357730-2 2008 To investigate the effect of sophocarpine (SC) on HERG channel stably expressing in human embryonic kidney-293 (HEK293) cells, whole-cell patch-clamp technique was used to record HERG current and kinetic curves. sophocarpine 29-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 18357730-2 2008 To investigate the effect of sophocarpine (SC) on HERG channel stably expressing in human embryonic kidney-293 (HEK293) cells, whole-cell patch-clamp technique was used to record HERG current and kinetic curves. sophocarpine 43-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). sparfloxacin 149-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). sparfloxacin 149-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). telithromycin 177-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). telithromycin 177-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Moxifloxacin 191-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Moxifloxacin 191-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Erythromycin 204-216 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 18074444-3 2007 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Erythromycin 204-216 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 18074444-10 2007 Additional experiments with grepafloxacin indicate that the rank order to detect grepafloxacin-induced long QT was the wedge preparation>the Purkinje fiber>HERG>the isolated heart, where the isolated heart was unable to detect grepafloxacin-induced APD prolongation. grepafloxacin 81-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 18074444-10 2007 Additional experiments with grepafloxacin indicate that the rank order to detect grepafloxacin-induced long QT was the wedge preparation>the Purkinje fiber>HERG>the isolated heart, where the isolated heart was unable to detect grepafloxacin-induced APD prolongation. grepafloxacin 81-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 17826747-2 2007 The present study found that BAPTA-AM rapidly and reversibly suppressed human ether a-go-go-related gene (hERG or Kv11.1) K(+) current, human Kv1.3 and human Kv1.5 channel currents stably expressed in HEK 293 cells, and the effects were not related to Ca(2+) chelation. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 29-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-120 18057881-0 2008 The additive effects of the active component of grapefruit juice (naringenin) and antiarrhythmic drugs on HERG inhibition. naringenin 66-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 18057881-1 2008 BACKGROUND: Grapefruit juice causes significant QT prolongation in healthy volunteers and naringenin has been identified as the most potent human ether-a-go-go-related gene (HERG) channel blocker among several dietary flavonoids. naringenin 90-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 18057881-7 2008 Naringenin blocked HERG current dose dependently with an IC(50) of 173.3 +/- 3.1 microM. naringenin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 18057881-8 2008 Naringenin 100 microM alone inhibited HERG current by 31 +/- 6%, and this inhibitory effect was increased with coadministration of 1 or 10 microM antiarrhythmic drugs. naringenin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 18057881-9 2008 When 100 microM naringenin was added to antiarrhythmic drugs, greater HERG inhibition was demonstrated, compared to the current inhibition caused by antiarrhythmic drugs alone. naringenin 16-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 18057881-11 2008 CONCLUSIONS: There is an additive inhibitory effect on HERG current when naringenin is combined with I(Kr)-blocking antiarrhythmic drugs. naringenin 73-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 18057887-2 2008 Our previous studies have shown that extracellular acidosis and hyperkalemia attenuate the HERG-inhibitory effect of proarrhythmic drugs, e.g. quinidine, but have little impact on the less-proarrhythmic drug amiodarone. Amiodarone 208-218 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 18057887-5 2008 Our results showed that ibutilide was a potent HERG inhibitor. ibutilide 24-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 18057887-10 2008 CONCLUSION: Extracellular acidosis and hyperkalemia attenuate the HERG-inhibitory effect of ibutilide. ibutilide 92-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 18057887-11 2008 The differences in HERG inhibition between acidic and hyperkalemic regions compared to normal regions in the myocardium may result in heterogeneity in repolarization, which may contribute to the proarrhythmic toxicity of ibutilide. ibutilide 221-230 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 17711440-0 2007 In vivo effects of mutant HERG K+ channel inhibition by disopyramide in patients with a short QT-1 syndrome: a pilot study. Disopyramide 56-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 17965852-0 2007 Anticholinergic antiparkinson drug orphenadrine inhibits HERG channels: block attenuation by mutations of the pore residues Y652 or F656. Orphenadrine 35-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 17965852-5 2007 Orphenadrine inhibits cloned HERG channels in a concentration dependent manner, yielding an IC(50) of 0.85 microM in HEK cells. Orphenadrine 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 17965852-11 2007 In conclusion, we show that the anticholinergic agent orphenadrine is an antagonist at HERG channels. Orphenadrine 54-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 17711440-1 2007 INTRODUCTION: Quinidine has been evaluated in patients with a short QT-1 syndrome caused by an IKr gain-of-function mutation of HERG. Quinidine 14-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 17711440-3 2007 The aim of the present study was to test the in vivo effects of disopyramide in patients with short QT-1 syndrome caused by a N588K mutation in HERG. Disopyramide 64-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 144-148 17448117-5 2007 In this article, we demonstrate that ERG1-2 channels are also reversibly inhibited by XE991 in the micromolar range (EC(50) 107 microM for ERG1). 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone 86-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-43 17768047-0 2007 Discovery of novel and orally active NR2B-selective N-methyl-D-aspartate (NMDA) antagonists, pyridinol derivatives with reduced HERG binding affinity. N-Methylaspartate 52-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 17768047-0 2007 Discovery of novel and orally active NR2B-selective N-methyl-D-aspartate (NMDA) antagonists, pyridinol derivatives with reduced HERG binding affinity. N-Methylaspartate 74-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 17768047-0 2007 Discovery of novel and orally active NR2B-selective N-methyl-D-aspartate (NMDA) antagonists, pyridinol derivatives with reduced HERG binding affinity. 2-hydroxypyridine 93-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 17768047-2 2007 Structure-activity relationship investigation led to N-[cis-4-hydroxy-4-(5-hydroxypyridin-2-yl)cyclohexyl]-3-henylpropanamide as an orally active NR2B-subtype selective N-methyl-D-aspartate (NMDA) receptor antagonist with very weak HERG (human ether-a-go-go related gene) binding (IC(50)> 30 microM). n-[cis-4-hydroxy-4-(5-hydroxypyridin-2-yl)cyclohexyl 53-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-236 17768047-2 2007 Structure-activity relationship investigation led to N-[cis-4-hydroxy-4-(5-hydroxypyridin-2-yl)cyclohexyl]-3-henylpropanamide as an orally active NR2B-subtype selective N-methyl-D-aspartate (NMDA) receptor antagonist with very weak HERG (human ether-a-go-go related gene) binding (IC(50)> 30 microM). henylpropanamide 109-125 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-236 17763876-9 2007 Hydrogen peroxide was shown to increase HERG1a K+ current in heterologous expression systems, which constitutes an apoptotic signal. Hydrogen Peroxide 0-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-45 17622552-0 2007 Phosphatidylinositol 4,5-bisphosphate interactions with the HERG K(+) channel. Phosphatidylinositol 4,5-Diphosphate 0-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 17622552-6 2007 A recently described pathway for regulation of HERG is through channel interaction with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2). phospholipid phosphatidylinositol 4,5-bisphosphate 92-142 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 17622552-6 2007 A recently described pathway for regulation of HERG is through channel interaction with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2). Phosphatidylinositol 4,5-Diphosphate 144-148 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 17624316-3 2007 Functional consequences of oxidative modification of methionine in human ether a go-go related gene 1 (hERG1), which encodes cardiac I(Kr) channels, are unknown. Methionine 53-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-108 17171344-5 2007 Screening for SCN5A and HERG candidate genes identified a heterozygous missense mutation 1810G-->A in exon 7 of HERG that leads to the substitution of the amino acid glycine by serine (G604S); this mutation was located in the S5/pore region of the HERG protein and was associated with a malignant phenotype. Glycine 169-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 17171344-5 2007 Screening for SCN5A and HERG candidate genes identified a heterozygous missense mutation 1810G-->A in exon 7 of HERG that leads to the substitution of the amino acid glycine by serine (G604S); this mutation was located in the S5/pore region of the HERG protein and was associated with a malignant phenotype. Glycine 169-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 17171344-5 2007 Screening for SCN5A and HERG candidate genes identified a heterozygous missense mutation 1810G-->A in exon 7 of HERG that leads to the substitution of the amino acid glycine by serine (G604S); this mutation was located in the S5/pore region of the HERG protein and was associated with a malignant phenotype. Glycine 169-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 17171344-5 2007 Screening for SCN5A and HERG candidate genes identified a heterozygous missense mutation 1810G-->A in exon 7 of HERG that leads to the substitution of the amino acid glycine by serine (G604S); this mutation was located in the S5/pore region of the HERG protein and was associated with a malignant phenotype. Serine 180-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 17171344-5 2007 Screening for SCN5A and HERG candidate genes identified a heterozygous missense mutation 1810G-->A in exon 7 of HERG that leads to the substitution of the amino acid glycine by serine (G604S); this mutation was located in the S5/pore region of the HERG protein and was associated with a malignant phenotype. Serine 180-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 17171344-5 2007 Screening for SCN5A and HERG candidate genes identified a heterozygous missense mutation 1810G-->A in exon 7 of HERG that leads to the substitution of the amino acid glycine by serine (G604S); this mutation was located in the S5/pore region of the HERG protein and was associated with a malignant phenotype. Serine 180-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 17092964-5 2007 Experiments in HERG-transfected (HERG: human ether-a-gogo-related gene) HEK293 cells (n = 36) demonstrated concentration-dependent block of the rapid component (I(Kr)) of the delayed rectifier potassium current: tail current was decreased 50% at olanzapine 3.8 microM. Olanzapine 246-256 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 17092964-5 2007 Experiments in HERG-transfected (HERG: human ether-a-gogo-related gene) HEK293 cells (n = 36) demonstrated concentration-dependent block of the rapid component (I(Kr)) of the delayed rectifier potassium current: tail current was decreased 50% at olanzapine 3.8 microM. Olanzapine 246-256 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 17560554-0 2007 Inhibition of the HERG potassium channel by the tricyclic antidepressant doxepin. Doxepin 73-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 17560554-2 2007 This study investigated the effects on HERG channels of doxepin, a tricyclic antidepressant linked to QT interval prolongation and cardiac arrhythmia. Doxepin 56-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 17560554-4 2007 Doxepin inhibited I(HERG) with an IC(50) value of 6.5+/-1.4 microM and native I(Kr) with an IC(50) of 4.4+/-0.6 microM. Doxepin 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 17560554-8 2007 HERG channel blockade is likely to underpin reported cases of QT interval prolongation with doxepin. Doxepin 92-99 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 17560554-9 2007 Notably, this study also establishes doxepin as an effective inhibitor of mutant (N588K) HERG channels responsible for variant 1 of the short QT syndrome. Doxepin 37-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 17785939-4 2007 Mitemcinal and its metabolites, GM-577 and GM-625, inhibited the human ether-a-go-go-related gene (HERG) tail current in a concentration-dependent manner with IC(50) values of 20.2, 41.7, and 55.0 microM, respectively. mitemcinal 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 17785939-4 2007 Mitemcinal and its metabolites, GM-577 and GM-625, inhibited the human ether-a-go-go-related gene (HERG) tail current in a concentration-dependent manner with IC(50) values of 20.2, 41.7, and 55.0 microM, respectively. gm-577 32-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 17610913-5 2007 The compound NS1643 has earlier been reported as an ERG1 channel activator. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 13-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 17610913-6 2007 We found that NS1643 also activates the ERG2 channel; however, the molecular mechanism of the activation differs between the ERG1 and ERG2 channels. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 14-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). Cholesterol 22-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-69 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). Cholesterol 22-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-238 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). Cholesterol 22-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-238 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). methyl-beta-cyclodextrin 85-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-69 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). methyl-beta-cyclodextrin 85-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-238 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). methyl-beta-cyclodextrin 85-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-238 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). methyl-beta-cyclodextrin 111-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-69 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). methyl-beta-cyclodextrin 111-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-238 18708743-6 2007 Depletion of membrane cholesterol from HEK293 cells expressing Kv11.1 channels using methyl-beta-cyclodextrin (MbetaCD) caused a positive shift of the voltage dependence of activation and an acceleration of deactivation kinetics of Kv11.1 current (I(Kv11.1)). methyl-beta-cyclodextrin 111-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 232-238 18708743-7 2007 Cholesterol loading of HEK293 cells reduced the steep voltage dependence of I(Kv11.1) activation and accelerated the inactivation kinetics of I(Kv11.1). Cholesterol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-84 18708743-7 2007 Cholesterol loading of HEK293 cells reduced the steep voltage dependence of I(Kv11.1) activation and accelerated the inactivation kinetics of I(Kv11.1). Cholesterol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 144-150 18708743-9 2007 We conclude that Kv11.1 protein localizes in cholesterol and sphingolipid enriched membranes and that membrane cholesterol can modulate I(Kv11.1) and I(Kr). Cholesterol 45-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-23 18708743-9 2007 We conclude that Kv11.1 protein localizes in cholesterol and sphingolipid enriched membranes and that membrane cholesterol can modulate I(Kv11.1) and I(Kr). Sphingolipids 61-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-23 18708743-9 2007 We conclude that Kv11.1 protein localizes in cholesterol and sphingolipid enriched membranes and that membrane cholesterol can modulate I(Kv11.1) and I(Kr). Cholesterol 111-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-23 18708743-9 2007 We conclude that Kv11.1 protein localizes in cholesterol and sphingolipid enriched membranes and that membrane cholesterol can modulate I(Kv11.1) and I(Kr). Cholesterol 111-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 138-144 17610913-9 2007 In contrast, for ERG1, NS1643 causes a right-ward shift in the voltage-dependent release from inactivation but does not affect time-constants of deactivation. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 23-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 17610913-10 2007 Because of these differences in the responses of ERG1 and ERG2 to NS1643, NS1643 can be used as a pharmacological tool to address ERG channel function. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 74-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 18708743-0 2007 Kv11.1 (ERG1) K+ channels localize in cholesterol and sphingolipid enriched membranes and are modulated by membrane cholesterol. Cholesterol 38-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-6 18708743-0 2007 Kv11.1 (ERG1) K+ channels localize in cholesterol and sphingolipid enriched membranes and are modulated by membrane cholesterol. Cholesterol 38-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 8-12 18708743-0 2007 Kv11.1 (ERG1) K+ channels localize in cholesterol and sphingolipid enriched membranes and are modulated by membrane cholesterol. Sphingolipids 54-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-6 18708743-0 2007 Kv11.1 (ERG1) K+ channels localize in cholesterol and sphingolipid enriched membranes and are modulated by membrane cholesterol. Sphingolipids 54-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 8-12 18708743-0 2007 Kv11.1 (ERG1) K+ channels localize in cholesterol and sphingolipid enriched membranes and are modulated by membrane cholesterol. Cholesterol 116-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-6 18708743-0 2007 Kv11.1 (ERG1) K+ channels localize in cholesterol and sphingolipid enriched membranes and are modulated by membrane cholesterol. Cholesterol 116-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 8-12 18708743-3 2007 Immunocytochemistry and membrane fractionation using discontinuous sucrose density gradients of adult canine ventricular tissue showed that Kv11.1 channel protein localized to both the cell surface and T-tubular sarcolemma. Sucrose 67-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 140-146 18708743-4 2007 Furthermore, density gradient membrane fractionation using detergent (Triton X-100) and non-detergent (OptiPrep) methods from canine ventricular myocytes or HEK293 cells demonstrated that Kv11.1 protein, along with MiRP1 and Kv7.1 (KCNQ1) proteins, localize in cholesterol and sphingolipid enriched membrane fractions. Cholesterol 261-272 potassium voltage-gated channel subfamily H member 2 Homo sapiens 188-194 18708743-4 2007 Furthermore, density gradient membrane fractionation using detergent (Triton X-100) and non-detergent (OptiPrep) methods from canine ventricular myocytes or HEK293 cells demonstrated that Kv11.1 protein, along with MiRP1 and Kv7.1 (KCNQ1) proteins, localize in cholesterol and sphingolipid enriched membrane fractions. Sphingolipids 277-289 potassium voltage-gated channel subfamily H member 2 Homo sapiens 188-194 18708743-5 2007 In HEK293 cells, Kv11.1 channels, but not long QT-associated mutant G601S-Kv11.1 channels, also localized to cholesterol and sphingolipid enriched membrane fractions. Cholesterol 109-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-23 18708743-5 2007 In HEK293 cells, Kv11.1 channels, but not long QT-associated mutant G601S-Kv11.1 channels, also localized to cholesterol and sphingolipid enriched membrane fractions. Sphingolipids 125-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-23 17460149-10 2007 In HERG-transfected COS-7 cells, SM-21 potentiated the ifenprodil-induced blockade of the HERG current. SM 21 33-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 17460149-10 2007 In HERG-transfected COS-7 cells, SM-21 potentiated the ifenprodil-induced blockade of the HERG current. SM 21 33-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 17460149-10 2007 In HERG-transfected COS-7 cells, SM-21 potentiated the ifenprodil-induced blockade of the HERG current. ifenprodil 55-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 17460149-10 2007 In HERG-transfected COS-7 cells, SM-21 potentiated the ifenprodil-induced blockade of the HERG current. ifenprodil 55-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 17641836-2 2007 The functional role of HERG1 K+ channels in leukemic cell proliferation was measured by MTT assay, and cell cycle and apoptosis were analyzed by flow cytometry. monooxyethylene trimethylolpropane tristearate 88-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-28 17641836-5 2007 In addition, leukemic cell proliferation was dramatically inhibited by HERG K+ channel special inhibitor E-4031. E 4031 105-111 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 17448117-5 2007 In this article, we demonstrate that ERG1-2 channels are also reversibly inhibited by XE991 in the micromolar range (EC(50) 107 microM for ERG1). 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone 86-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 18690032-0 2007 Mutational analysis of block and facilitation of HERG current by a class III anti-arrhythmic agent, nifekalant. nifekalant 100-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 17457128-2 2007 Although droperidol potently inhibits human ether-a-go-go-related gene (HERG) channels, the molecular mode of this interaction is unknown. Droperidol 9-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 17457128-5 2007 METHODS: Molecular mechanisms of HERG current inhibition by droperidol were established using two-electrode voltage clamp recordings of Xenopus laevis oocytes expressing wild-type and mutant channels. Droperidol 60-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 17457128-8 2007 RESULTS: Droperidol inhibited currents through HERG wild-type channels with a concentration of half-maximal inhibition of 0.6-0.9 microM. Droperidol 9-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 17457128-14 2007 CONCLUSIONS: Droperidol is a high-affinity blocker of HERG channels. Droperidol 13-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 18690032-3 2007 Besides block, some chemicals such as the class III anti-arrhythmic agent nifekalant stimulate HERG at low potentials by shifting its activation curve towards hyperpolarizing voltages. nifekalant 74-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 95-99 18690032-6 2007 Alanine-scanning mutagenesis was performed in the pore region of HERG. Alanine 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 17497253-9 2007 Pharmacological treatment has been studied and it could be demonstrated, that some mutant currents may be insufficiently suppressed by drugs targeted to block the specific current such as, e.g., sotalol or ibutilide in patients with a mutation in the IKr-coding gene KCNH2 (HERG). Sotalol 195-202 potassium voltage-gated channel subfamily H member 2 Homo sapiens 267-272 17497253-9 2007 Pharmacological treatment has been studied and it could be demonstrated, that some mutant currents may be insufficiently suppressed by drugs targeted to block the specific current such as, e.g., sotalol or ibutilide in patients with a mutation in the IKr-coding gene KCNH2 (HERG). Sotalol 195-202 potassium voltage-gated channel subfamily H member 2 Homo sapiens 274-278 17497253-9 2007 Pharmacological treatment has been studied and it could be demonstrated, that some mutant currents may be insufficiently suppressed by drugs targeted to block the specific current such as, e.g., sotalol or ibutilide in patients with a mutation in the IKr-coding gene KCNH2 (HERG). ibutilide 206-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 267-272 17497253-9 2007 Pharmacological treatment has been studied and it could be demonstrated, that some mutant currents may be insufficiently suppressed by drugs targeted to block the specific current such as, e.g., sotalol or ibutilide in patients with a mutation in the IKr-coding gene KCNH2 (HERG). ibutilide 206-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 274-278 17376289-0 2007 Inhibitory effects of coronary vasodilator papaverine on heterologously-expressed HERG currents in Xenopus oocytes. Papaverine 43-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 17376289-1 2007 AIM: To characterize the effects of papaverine on HERG channels expressed in Xenopus oocytes as well as cardiac action potential in rabbit ventricular myocytes. Papaverine 36-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 17376289-6 2007 The blockade of papaverine on HERG currents was not voltage-dependent. Papaverine 16-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 17376289-7 2007 The slope conductance measured as a slope of the fully activated HERG current-voltage curves decreased from 78.03+/-4.25 muS of the control to 56.84+/-5.33, 36.06+/-6.53, and 27.09+/-5.50 microS (n=4) by 30, 100, and 300 micromol/L of papaverine, respectively. Papaverine 235-245 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 17376289-9 2007 Papaverine blocked HERG channels in the closed, open, and inactivated states. Papaverine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 17376289-10 2007 CONCLUSION: These results showed that papaverine blocked HERG channels in a voltage- and state-independent manner, which may most likely be the major mechanism of papaverine-induced cardiac arrhythmia reported in humans. Papaverine 38-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 17376289-10 2007 CONCLUSION: These results showed that papaverine blocked HERG channels in a voltage- and state-independent manner, which may most likely be the major mechanism of papaverine-induced cardiac arrhythmia reported in humans. Papaverine 163-173 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 17189275-5 2007 KCR1 and the yeast alpha-1,2-glucosyltransferase ALG10 exhibit sequence homology, and like KCR1, ALG10 diminished HERG block by dofetilide. dofetilide 128-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 17489361-0 2007 Maprotiline block of the human ether-a-go-go-related gene (HERG) K+ channel. Maprotiline 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 17489361-2 2007 We studied the effects of maprotiline on human ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes and HEK293 cells. Maprotiline 26-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-79 17489361-3 2007 Maprotiline induced a concentration-dependent decrease in current amplitudes at the end of the voltage steps and tail currents of HERG. Maprotiline 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 17489361-5 2007 The IC50 for a maprotiline block of HERG current in Xenopus oocytes did not change according to depolarization; 39.5 +/- 3.2 microM at -40 mV and 43.6 +/- 2.8 microM at +40 mV. Maprotiline 15-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 17489361-6 2007 The block of HERG by maprotiline was examined after treatment of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent that neutralizes the positively charged amino-groups of peptide N-terminal and lysine residues. Maprotiline 21-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 17489361-6 2007 The block of HERG by maprotiline was examined after treatment of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent that neutralizes the positively charged amino-groups of peptide N-terminal and lysine residues. Trinitrobenzenesulfonic Acid 96-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 17489361-6 2007 The block of HERG by maprotiline was examined after treatment of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent that neutralizes the positively charged amino-groups of peptide N-terminal and lysine residues. Lysine 205-211 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 17489361-8 2007 The IC50 for the maprotiline-induced blockade of HERG currents in HEK293 cells at 36 degrees C was 0.13 microM at +20 mV. Maprotiline 17-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 17489361-9 2007 Our findings suggest that the arrhythmogenic side effects of maprotiline are caused by a blockade of HERG and possibly by a blockade of delayed rectifier K+ channel. Maprotiline 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 17189275-8 2007 HERG itself is not the target for KCR1-mediated glycosylation because the dofetilide response of glycosylation-deficient HERG(N598Q) was still modulated by KCR1. dofetilide 74-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 17189275-8 2007 HERG itself is not the target for KCR1-mediated glycosylation because the dofetilide response of glycosylation-deficient HERG(N598Q) was still modulated by KCR1. dofetilide 74-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 17518040-0 2007 [Effects of matrine, oxymatrine and resveratrol on HERG channel expression]. matrine 12-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 17325511-1 2007 BACKGROUND: Local anesthetics interact with human ether-a-go-go-related gene (HERG) channels via the aromatic amino acids Y652 and F656 in the S6 region. Amino Acids, Aromatic 101-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 17325511-2 2007 This study aimed to establish whether the residues T623, S624, and V625 residing deeper within the pore are also involved in HERG channel block by bupivacaine. Bupivacaine 147-158 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 17325511-8 2007 Inhibition of HERG tail currents by bupivacaine (300 microm) was reduced by all mutations (P < 0.001). Bupivacaine 36-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 17325511-11 2007 CONCLUSIONS: The authors" results indicate that not only the aromatic residues Y652 and F656 but also residues residing deeper within the pore and close to the selectivity filter of HERG channels are involved in inhibition of HERG channels by the low-affinity blocker bupivacaine. Bupivacaine 268-279 potassium voltage-gated channel subfamily H member 2 Homo sapiens 182-186 17325511-11 2007 CONCLUSIONS: The authors" results indicate that not only the aromatic residues Y652 and F656 but also residues residing deeper within the pore and close to the selectivity filter of HERG channels are involved in inhibition of HERG channels by the low-affinity blocker bupivacaine. Bupivacaine 268-279 potassium voltage-gated channel subfamily H member 2 Homo sapiens 226-230 17109852-3 2007 The acute effects of the novel prokinetic prucalopride were examined on heterologously expressed HERG channels in human embryonic kidney (HEK) 293 cells using the whole-cell patch-clamp technique. prucalopride 42-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 17109852-4 2007 Prucalopride inhibited HERG channels in a concentration-dependent manner with an IC(50) of 4.1 microM. prucalopride 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 17109852-8 2007 Though prucalopride blocks HERG channels this is unlikely to be significant at clinically relevant concentrations. prucalopride 7-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 17762170-0 2007 Sphingolipid metabolite ceramide causes metabolic perturbation contributing to HERG K+ channel dysfunction. Sphingolipids 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 17762170-0 2007 Sphingolipid metabolite ceramide causes metabolic perturbation contributing to HERG K+ channel dysfunction. Ceramides 24-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 17762170-3 2007 To investigate how ceramide is involved in modulating cardiac repolarization, we performed whole-cell patch-clamp studies on HERG current (I(HERG)), a critical determinant of cardiac repolarization, expressed in HEK293 cells. Ceramides 19-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 17762170-11 2007 The inhibitory effect of C2 on I(HERG) was reversed by antioxidants vitamin E or MnTBAP. Vitamin E 68-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 17762170-13 2007 We conclude that ceramide depresses I(HERG) mainly via ROS overproduction and ceramide-induced I(HERG) impairment may contribute to QT prolongation in prolonged myocardial ischemia, heart failure and diabetic cardiomyopathy. Ceramides 17-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 17762170-13 2007 We conclude that ceramide depresses I(HERG) mainly via ROS overproduction and ceramide-induced I(HERG) impairment may contribute to QT prolongation in prolonged myocardial ischemia, heart failure and diabetic cardiomyopathy. Reactive Oxygen Species 55-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 16938156-0 2007 Inhibition of HERG channels by the local anaesthetic articaine. Carticaine 53-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 16938156-2 2007 Human ether-a-go-go-related gene (HERG) potassium channels constitute potential targets involved in cardiotoxic side-effects of various pharmacological agents including amide local anaesthetics. Amides 169-174 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 16938156-3 2007 The aim of this study was to determine the sensitivity of HERG channels to the inhibitory action of articaine and to further evaluate the effect of the mutations Y652A and F656A in the putative drug-binding region of HERG on the sensitivity for articaine. Carticaine 100-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 16938156-3 2007 The aim of this study was to determine the sensitivity of HERG channels to the inhibitory action of articaine and to further evaluate the effect of the mutations Y652A and F656A in the putative drug-binding region of HERG on the sensitivity for articaine. Carticaine 100-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 217-221 16938156-3 2007 The aim of this study was to determine the sensitivity of HERG channels to the inhibitory action of articaine and to further evaluate the effect of the mutations Y652A and F656A in the putative drug-binding region of HERG on the sensitivity for articaine. Carticaine 245-254 potassium voltage-gated channel subfamily H member 2 Homo sapiens 217-221 16938156-4 2007 METHODS: We examined the inhibition of wild-type and mutant HERG channels, transiently expressed in Chinese hamster ovary cells by articaine. Carticaine 131-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 16938156-6 2007 RESULTS: Inhibition of HERG wild-type and HERG Y652A channels by articaine was concentration dependent and reversible. Carticaine 65-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 16938156-6 2007 RESULTS: Inhibition of HERG wild-type and HERG Y652A channels by articaine was concentration dependent and reversible. Carticaine 65-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 16960444-9 2007 Amiodarone, azimilide, dofetilide, quinidine and sotalol all produced a dose-dependent inhibition of HERG current. Amiodarone 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16960444-9 2007 Amiodarone, azimilide, dofetilide, quinidine and sotalol all produced a dose-dependent inhibition of HERG current. azimilide 12-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16960444-9 2007 Amiodarone, azimilide, dofetilide, quinidine and sotalol all produced a dose-dependent inhibition of HERG current. dofetilide 23-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16960444-9 2007 Amiodarone, azimilide, dofetilide, quinidine and sotalol all produced a dose-dependent inhibition of HERG current. Quinidine 35-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16960444-9 2007 Amiodarone, azimilide, dofetilide, quinidine and sotalol all produced a dose-dependent inhibition of HERG current. Sotalol 49-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16960444-12 2007 Raising the extracellular potassium to 10 mmol/l, HERG block by azimilide, dofetilide, quinidine and sotalol was significantly decreased, while the block by amiodarone was unchanged. Potassium 26-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 16960444-12 2007 Raising the extracellular potassium to 10 mmol/l, HERG block by azimilide, dofetilide, quinidine and sotalol was significantly decreased, while the block by amiodarone was unchanged. azimilide 64-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 16960444-12 2007 Raising the extracellular potassium to 10 mmol/l, HERG block by azimilide, dofetilide, quinidine and sotalol was significantly decreased, while the block by amiodarone was unchanged. dofetilide 75-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 16960444-12 2007 Raising the extracellular potassium to 10 mmol/l, HERG block by azimilide, dofetilide, quinidine and sotalol was significantly decreased, while the block by amiodarone was unchanged. Quinidine 87-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 16960444-12 2007 Raising the extracellular potassium to 10 mmol/l, HERG block by azimilide, dofetilide, quinidine and sotalol was significantly decreased, while the block by amiodarone was unchanged. Sotalol 101-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 17408310-14 2007 Cisapride was found to bind the human ether-a-go-go-related gene (HERG) potassium channel, which provides a plausible mechanism for QTc interval prolongation/arrhythmia. Cisapride 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 17408310-15 2007 Other QTc interval-prolonging/arrhythmic drugs that also bind to HERG provided an analogy for cisapride causing QTc interval prolongation/arrhythmia via this mechanism. Cisapride 94-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 17518040-0 2007 [Effects of matrine, oxymatrine and resveratrol on HERG channel expression]. oxymatrine 21-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 17518040-0 2007 [Effects of matrine, oxymatrine and resveratrol on HERG channel expression]. Resveratrol 36-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 17518040-1 2007 Because HERG potassium channel has important effects on both proarrhythmia and antiarrhythmia, we use immunofluorescence and Western blotting methods to detect the expression of HERG channel of HERG-HEK cells in different concentrations of matrine, oxymatrine and resveratrol. matrine 240-247 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 17518040-1 2007 Because HERG potassium channel has important effects on both proarrhythmia and antiarrhythmia, we use immunofluorescence and Western blotting methods to detect the expression of HERG channel of HERG-HEK cells in different concentrations of matrine, oxymatrine and resveratrol. matrine 240-247 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 17518040-1 2007 Because HERG potassium channel has important effects on both proarrhythmia and antiarrhythmia, we use immunofluorescence and Western blotting methods to detect the expression of HERG channel of HERG-HEK cells in different concentrations of matrine, oxymatrine and resveratrol. oxymatrine 249-259 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 17518040-1 2007 Because HERG potassium channel has important effects on both proarrhythmia and antiarrhythmia, we use immunofluorescence and Western blotting methods to detect the expression of HERG channel of HERG-HEK cells in different concentrations of matrine, oxymatrine and resveratrol. oxymatrine 249-259 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 17518040-1 2007 Because HERG potassium channel has important effects on both proarrhythmia and antiarrhythmia, we use immunofluorescence and Western blotting methods to detect the expression of HERG channel of HERG-HEK cells in different concentrations of matrine, oxymatrine and resveratrol. Resveratrol 264-275 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 17518040-1 2007 Because HERG potassium channel has important effects on both proarrhythmia and antiarrhythmia, we use immunofluorescence and Western blotting methods to detect the expression of HERG channel of HERG-HEK cells in different concentrations of matrine, oxymatrine and resveratrol. Resveratrol 264-275 potassium voltage-gated channel subfamily H member 2 Homo sapiens 178-182 17518040-2 2007 The findings showed that both matrine (1 micromol x L(-1) ) and oxymatrine ( 1micromol x L (-1) ) increased HERG channel expression ( n = 5, P < 0. matrine 30-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 17518040-2 2007 The findings showed that both matrine (1 micromol x L(-1) ) and oxymatrine ( 1micromol x L (-1) ) increased HERG channel expression ( n = 5, P < 0. oxymatrine 64-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 17518040-3 2007 05 ) , while matrine (100 micromol x L(-1) ) decreased HERG channel expression ( n = 5, P < 0. matrine 13-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 17518040-5 2007 In conclusion, different concentrations of matrine and oxymatrine affect HERG channel expression, while there is no relationship between resveratrol and HERG channel expression. oxymatrine 55-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 17313833-0 2006 [Erythromycin inhibits the proliferation of HERG K+ channel highly expressing cancer cells and shows synergy with anticancer drugs]. Erythromycin 1-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). telithromycin 177-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Moxifloxacin 191-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Moxifloxacin 191-203 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Erythromycin 204-216 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). Erythromycin 204-216 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 17054943-10 2006 Additional experiments with grepafloxacin indicate that the rank order to detect grepafloxacin-induced long QT was the wedge preparation>the Purkinje fiber>HERG>the isolated heart, where the isolated heart was unable to detect grepafloxacin-induced APD prolongation. grepafloxacin 81-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 17054943-10 2006 Additional experiments with grepafloxacin indicate that the rank order to detect grepafloxacin-induced long QT was the wedge preparation>the Purkinje fiber>HERG>the isolated heart, where the isolated heart was unable to detect grepafloxacin-induced APD prolongation. grepafloxacin 81-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 17313833-1 2006 OBJECTIVE: To investigate the effects of erythromycin on the proliferation of the human ether-a-go-go related gene (HERG) K(+) channel highly expressing cancer cells and its synergy with antitumor chemotherapeutic agents. Erythromycin 41-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 116-120 17313833-18 2006 CONCLUSION: Erythromycin inhibits the proliferation and induces the apoptosis of cancer cells with high HERG K(+) channel expression. Erythromycin 12-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 17027138-3 2006 Sparfloxacin blocked HERG currents half-maximally (IC(50) value) at a concentration of 33.2 microM, whereas norfloxacin and lomefloxacin each tested at a concentration of 300 microM inhibited HERG currents only by 2.8+/-3.6% and 12.3+/-4.7%, respectively. sparfloxacin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 17027138-3 2006 Sparfloxacin blocked HERG currents half-maximally (IC(50) value) at a concentration of 33.2 microM, whereas norfloxacin and lomefloxacin each tested at a concentration of 300 microM inhibited HERG currents only by 2.8+/-3.6% and 12.3+/-4.7%, respectively. Norfloxacin 108-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 192-196 17027138-3 2006 Sparfloxacin blocked HERG currents half-maximally (IC(50) value) at a concentration of 33.2 microM, whereas norfloxacin and lomefloxacin each tested at a concentration of 300 microM inhibited HERG currents only by 2.8+/-3.6% and 12.3+/-4.7%, respectively. lomefloxacin 124-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 192-196 17027138-7 2006 The amino group at position C(5) seems to be primarily responsible for the strong HERG current blocking property of sparfloxacin. sparfloxacin 116-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 16942825-7 2006 Quinidine proved to be efficient in prolonging the QT interval and rendering ventricular tachyarrhythmias non-inducible in patients with a mutation in KCNH2 (HERG). Quinidine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-156 16949586-3 2006 F468C mutation increased IC(50) for astemizole and imipramine but in contrast to HERG channels, only slightly for dofetilide. dofetilide 114-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 16647228-5 2006 To further investigate the electrophysiological basis of the arrhythmogenic potential of chloroform, we analysed inhibitory effects of chloroform on cloned HERG potassium channels, heterologously expressed in Xenopus oocytes and in Human Embryonic Kidney (HEK 293) cells using the double-electrode voltage-clamp technique and the whole-cell patch-clamp technique, respectively. Chloroform 135-145 potassium voltage-gated channel subfamily H member 2 Homo sapiens 156-160 16647228-6 2006 In HEK cells, chloroform blocked HERG tail currents with an IC(50) of 4.97mM. Chloroform 14-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 16647228-11 2006 Chloroform dependent block of HERG channels was voltage dependent with a decrease of inhibition at more positive membrane potentials. Chloroform 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 16647228-13 2006 In summary, chloroform blocked HERG potassium channels probably in a toxicologically relevant concentration. Chloroform 12-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). sparfloxacin 149-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). sparfloxacin 149-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 17054943-3 2006 Using the patch clamp technique on the human ether-a-gogo-related gene (HERG) current, the rank order for blockade of the HERG-current (IC(50)) was: sparfloxacin (44 microM)>telithromycin=moxifloxacin=erythromycin (+/-100 microM). telithromycin 177-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 16842817-0 2006 Disopyramide is an effective inhibitor of mutant HERG K+ channels involved in variant 1 short QT syndrome. Disopyramide 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 16842817-2 2006 The use of implantable cardioverter defibrillators helps to protect SQTS patients from ventricular fibrillation; however, pharmacological treatments to normalise the QT interval are limited: thus far only quinidine has been found to be effective in a subset of patients, with the SQT1 variant. Quinidine 205-214 potassium voltage-gated channel subfamily H member 2 Homo sapiens 280-284 16842817-4 2006 We demonstrate here that the N588K-HERG mutation only slightly attenuates I(HERG) blockade by the Class Ia antiarrhythmic drug disopyramide (1.5-fold elevation of IC(50)), compared to quinidine (3.5-fold elevation of IC(50)) and the Class III antiarrhythmic drug E-4031 (11.5-fold elevation of IC(50)). Disopyramide 127-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 16842817-4 2006 We demonstrate here that the N588K-HERG mutation only slightly attenuates I(HERG) blockade by the Class Ia antiarrhythmic drug disopyramide (1.5-fold elevation of IC(50)), compared to quinidine (3.5-fold elevation of IC(50)) and the Class III antiarrhythmic drug E-4031 (11.5-fold elevation of IC(50)). Disopyramide 127-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 16842817-4 2006 We demonstrate here that the N588K-HERG mutation only slightly attenuates I(HERG) blockade by the Class Ia antiarrhythmic drug disopyramide (1.5-fold elevation of IC(50)), compared to quinidine (3.5-fold elevation of IC(50)) and the Class III antiarrhythmic drug E-4031 (11.5-fold elevation of IC(50)). Quinidine 184-193 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 16842817-4 2006 We demonstrate here that the N588K-HERG mutation only slightly attenuates I(HERG) blockade by the Class Ia antiarrhythmic drug disopyramide (1.5-fold elevation of IC(50)), compared to quinidine (3.5-fold elevation of IC(50)) and the Class III antiarrhythmic drug E-4031 (11.5-fold elevation of IC(50)). E 4031 263-269 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 16842817-5 2006 Thus, of the drugs studied to date, disopyramide is the one least affected by the SQT1 HERG mutation. Disopyramide 36-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 16842817-5 2006 Thus, of the drugs studied to date, disopyramide is the one least affected by the SQT1 HERG mutation. Disopyramide 36-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 16842817-6 2006 Disopyramide is associated with QT prolongation in normal use and our findings provide a rational basis for its evaluation as a treatment for SQT1. Disopyramide 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 16942825-7 2006 Quinidine proved to be efficient in prolonging the QT interval and rendering ventricular tachyarrhythmias non-inducible in patients with a mutation in KCNH2 (HERG). Quinidine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 158-162 16686685-4 2006 Class III agents like dofetilide, clofilium, and sotalol, which can all cause a drug-induced form of LQT2, significantly lengthen action potential duration at 50% and 90% repolarization in isolated rabbit Purkinje fibers, and can initiate the formation of early afterdepolarizations, and extra beats. dofetilide 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16647228-0 2006 In vitro modulation of HERG channels by organochlorine solvent trichlormethane as potential explanation for proarrhythmic effects of chloroform. Hydrocarbons, Chlorinated 40-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 16647228-0 2006 In vitro modulation of HERG channels by organochlorine solvent trichlormethane as potential explanation for proarrhythmic effects of chloroform. Chloroform 63-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 16647228-0 2006 In vitro modulation of HERG channels by organochlorine solvent trichlormethane as potential explanation for proarrhythmic effects of chloroform. Chloroform 133-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 16860311-0 2006 Blockade of HERG human K+ channel and IKr of guinea pig cardiomyocytes by prochlorperazine. Prochlorperazine 74-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 16860311-2 2006 We studied the effects of prochlorperazine on human ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes and also in the delayed rectifier K+ current of guinea pig cardiomyocytes. Prochlorperazine 26-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 16860311-3 2006 Prochlorperazine induced a concentration-dependent decrease in current amplitudes at the end of the voltage steps and tail currents of HERG. Prochlorperazine 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 135-139 16860311-4 2006 The IC50 for a prochlorperazine block of HERG current in Xenopus oocytes progressively decreased relative to the degree of depolarization, from 42.1 microM at -40 mV to 37.4 microM at 0 mV to 22.6 microM at +40 mV. Prochlorperazine 15-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 16860311-5 2006 The block of HERG by prochlorperazine was use-dependent, exhibiting a more rapid onset and a greater steady-state block at higher frequencies of activation, while there was partial relief of the block with reduced frequencies. Prochlorperazine 21-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 16860311-7 2006 Our findings suggest that the arrhythmogenic side effects of prochlorperazine are caused by a blockade of HERG and the rapid component of the delayed rectifier K+ current rather than by a blockade of the slow component. Prochlorperazine 61-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 16633353-0 2006 Blockade of HERG human K+ channels and IKr of guinea-pig cardiomyocytes by the antipsychotic drug clozapine. Clozapine 98-107 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 16633353-2 2006 To investigate the arrhythmogenic side effects of clozapine, we studied the impact of clozapine on human ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes and HEK293 cells, and on the delayed rectifier K(+) currents of guinea-pig cardiomyocytes. Clozapine 86-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 16633353-3 2006 Clozapine dose-dependently decreased the amplitudes of the currents at the end of voltage steps, and the tail currents of HERG. Clozapine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 16633353-8 2006 Our findings collectively suggest that blockade of HERG currents and I(Kr), but not I(Ks), may contribute to the arrhythmogenic side effects of clozapine. Clozapine 144-153 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Nitrogen 73-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Nitrogen 73-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Nitrogen 73-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Hydrogen 113-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Hydrogen 113-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Hydrogen 113-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Oxygen 145-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Oxygen 145-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Oxygen 145-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Benzene 383-390 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Benzene 383-390 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-2 2006 Our new model suggests three key interactions such that (1) a protonated nitrogen of the channel blocker forms a hydrogen bond with the carbonyl oxygen of HERG residue T623; (2) an aromatic moiety of the channel blocker makes a pi-pi interaction with the aromatic ring of HERG residue Y652; and (3) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Benzene 383-390 potassium voltage-gated channel subfamily H member 2 Homo sapiens 272-276 16616004-3 2006 The previous model assumes two interactions such that (1) a protonated nitrogen of the channel blocker forms a cation-pi interaction with the aromatic ring of HERG residue Y652; and (2) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Nitrogen 71-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 159-163 16616004-3 2006 The previous model assumes two interactions such that (1) a protonated nitrogen of the channel blocker forms a cation-pi interaction with the aromatic ring of HERG residue Y652; and (2) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Nitrogen 71-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 286-290 16616004-3 2006 The previous model assumes two interactions such that (1) a protonated nitrogen of the channel blocker forms a cation-pi interaction with the aromatic ring of HERG residue Y652; and (2) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Benzene 270-277 potassium voltage-gated channel subfamily H member 2 Homo sapiens 159-163 16616004-3 2006 The previous model assumes two interactions such that (1) a protonated nitrogen of the channel blocker forms a cation-pi interaction with the aromatic ring of HERG residue Y652; and (2) a hydrophobic group of the channel blocker forms a hydrophobic interaction with the benzene ring of HERG residue F656. Benzene 270-277 potassium voltage-gated channel subfamily H member 2 Homo sapiens 286-290 16686685-4 2006 Class III agents like dofetilide, clofilium, and sotalol, which can all cause a drug-induced form of LQT2, significantly lengthen action potential duration at 50% and 90% repolarization in isolated rabbit Purkinje fibers, and can initiate the formation of early afterdepolarizations, and extra beats. clofilium 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16686685-4 2006 Class III agents like dofetilide, clofilium, and sotalol, which can all cause a drug-induced form of LQT2, significantly lengthen action potential duration at 50% and 90% repolarization in isolated rabbit Purkinje fibers, and can initiate the formation of early afterdepolarizations, and extra beats. Sotalol 49-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 16542653-0 2006 Inhibition of the HERG K+ channel by the antifungal drug ketoconazole depends on channel gating and involves the S6 residue F656. Ketoconazole 57-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 16542653-1 2006 The mechanism of human ether-a-go-go-related gene (HERG) K+ channel blockade by the antifungal agent ketoconazole was investigated using patch-clamp recording from mammalian cell lines. Ketoconazole 101-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 16542653-2 2006 Ketoconazole inhibited whole-cell HERG current (IHERG) with a clinically relevant half-maximal inhibitory drug concentration (IC50) value of 1.7 microM. Ketoconazole 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 16542653-5 2006 Thus, ketoconazole accesses the HERG channel pore-cavity on channel gating, and the S6 residue F656 is an important determinant of ketoconazole binding. Ketoconazole 6-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 16681037-0 2006 Chemical modification of the human ether-a-go-go-related gene (HERG) K+ current by the amino-group reagent trinitrobenzene sulfonic acid. Trinitrobenzenesulfonic Acid 107-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 16681037-1 2006 We investigated the effects of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent, on the human ether-a-go-go-related gene (HERG) K+ channels expressed in Xenopus oocytes. Trinitrobenzenesulfonic Acid 31-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 134-138 16681037-1 2006 We investigated the effects of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent, on the human ether-a-go-go-related gene (HERG) K+ channels expressed in Xenopus oocytes. Trinitrobenzenesulfonic Acid 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 134-138 16681037-3 2006 External application of TNBS at 10 mM for 5 min irreversibly shifted the curves for currents at the end of the pulse and tail currents of HERG to a more negative potential and decreased the maximal amplitude of the I(tail) curve (I(tail,max)). Trinitrobenzenesulfonic Acid 24-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 138-142 16681037-5 2006 TNBS shifted the time constant curves of both activation and deactivation of the HERG current to a more hyperpolarized potential; TNBS"s effect was greater on channel opening than channel closing. Trinitrobenzenesulfonic Acid 0-4 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 16681037-5 2006 TNBS shifted the time constant curves of both activation and deactivation of the HERG current to a more hyperpolarized potential; TNBS"s effect was greater on channel opening than channel closing. Trinitrobenzenesulfonic Acid 130-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 16474415-3 2006 This study was conducted in order to characterise the inhibition of hERG current (I(hERG)) by moxifloxacin, and to determine the role in drug binding of the S6 aromatic amino-acid residues Tyr652 and Phe656. Moxifloxacin 94-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-89 16534005-8 2006 The median change in QT interval during low-dose epinephrine infusion was -23 ms in the gene-negative group, 78 ms in LQT1, -4 ms in LQT2, and -58 ms in LQT3. Epinephrine 49-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 16446155-0 2006 Erythromycin block of the HERG K+ channel: accessibility to F656 and Y652. Erythromycin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 16446155-3 2006 Here we used whole-cell patch-clamp recording at 37 degrees C from heterologously expressed HERG channels in a mammalian cell line to show that erythromycin either produces a rapid open-state-dependent HERG channel inhibition, or components of both open-state-dependent and closed-state-dependent inhibition. Erythromycin 144-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 16407206-0 2006 Chronic inhibition of cardiac Kir2.1 and HERG potassium channels by celastrol with dual effects on both ion conductivity and protein trafficking. celastrol 68-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 16506891-0 2006 Evaluation of the rubidium efflux assay for preclinical identification of HERG blockade. Rubidium 18-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 16314852-6 2006 Carvedilol (nonselective), propranolol (nonselective) and ICI 118551 (beta(2)-selective) inhibited HERG current in a concentration-dependent manner (IC(50) 0.51, 3.9 and 9.2 microM, respectively). Carvedilol 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 16314852-6 2006 Carvedilol (nonselective), propranolol (nonselective) and ICI 118551 (beta(2)-selective) inhibited HERG current in a concentration-dependent manner (IC(50) 0.51, 3.9 and 9.2 microM, respectively). Propranolol 27-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 16314852-6 2006 Carvedilol (nonselective), propranolol (nonselective) and ICI 118551 (beta(2)-selective) inhibited HERG current in a concentration-dependent manner (IC(50) 0.51, 3.9 and 9.2 microM, respectively). ICI 118551 58-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 16314852-9 2006 Inhibition of HERG current by carvedilol, propranolol and ICI 118551 was partially but significantly attenuated in Y652A and F656C mutant channels. Carvedilol 30-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 16314852-9 2006 Inhibition of HERG current by carvedilol, propranolol and ICI 118551 was partially but significantly attenuated in Y652A and F656C mutant channels. Propranolol 42-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 16278312-5 2006 Since two of the drugs that were ineffective I(HERG) blockers, arsenic trioxide and pentamidine, have been associated with cardiac repolarization delays (QT interval lengthening) and torsades de pointes ventricular arrhythmias in patients, we chose to evaluate them further using the isolated perfused rabbit heart model. Arsenic Trioxide 63-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 16278312-5 2006 Since two of the drugs that were ineffective I(HERG) blockers, arsenic trioxide and pentamidine, have been associated with cardiac repolarization delays (QT interval lengthening) and torsades de pointes ventricular arrhythmias in patients, we chose to evaluate them further using the isolated perfused rabbit heart model. Pentamidine 84-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 16480714-7 2006 The HERG-mediated potassium and the SCN5A-mediated sodium currents, however, were only slightly reduced by estradiol at concentrations of up to 30 microM. Potassium 18-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 16480714-7 2006 The HERG-mediated potassium and the SCN5A-mediated sodium currents, however, were only slightly reduced by estradiol at concentrations of up to 30 microM. Estradiol 107-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 16446155-3 2006 Here we used whole-cell patch-clamp recording at 37 degrees C from heterologously expressed HERG channels in a mammalian cell line to show that erythromycin either produces a rapid open-state-dependent HERG channel inhibition, or components of both open-state-dependent and closed-state-dependent inhibition. Erythromycin 144-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 202-206 16446155-4 2006 Alanine-substitution of HERG"s canonical determinants of blockade revealed that Y652 was not important as a molecular determinant of blockade, and that mutation of F656 resulted in only weak attenuation of inhibition. Alanine 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 16506891-3 2006 We evaluated the rubidium efflux assay for its ability to determine block of the hERG potassium channel. Rubidium 17-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 16291873-0 2006 Drug binding interactions in the inner cavity of HERG channels: molecular insights from structure-activity relationships of clofilium and ibutilide analogs. clofilium 124-133 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 16291873-0 2006 Drug binding interactions in the inner cavity of HERG channels: molecular insights from structure-activity relationships of clofilium and ibutilide analogs. ibutilide 138-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 16368812-3 2006 To clarify the mechanisms of anesthetics on HERG channels, we monitored the electrocardiogram and measured QT intervals in the guinea pig in the presence of sevoflurane and propofol. Propofol 173-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 16368812-7 2006 Sevoflurane (1%-4%), in a dose-dependent manner, inhibited the HERG outward tail currents (9.7%-26.6%), whereas steady-state currents were inhibited only at large concentrations. Sevoflurane 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 16368812-10 2006 In conclusion, compared with steady-state currents, sevoflurane was more potent in inhibiting the outward tail currents, suggesting that sevoflurane may modulate the HERG channel kinetics in its inactivated state. Sevoflurane 52-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 166-170 16368812-10 2006 In conclusion, compared with steady-state currents, sevoflurane was more potent in inhibiting the outward tail currents, suggesting that sevoflurane may modulate the HERG channel kinetics in its inactivated state. Sevoflurane 137-148 potassium voltage-gated channel subfamily H member 2 Homo sapiens 166-170 16472284-4 2006 DNA samples were genotyped for the nucleotide 2690 A>C variation of the KCNH2 gene, corresponding to the KCNH2 K(lysine)897T(threonine) amino acid polymorphism. Lysine 116-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-80 16423345-2 2006 Here we investigate the molecular mechanisms of voltage-dependent block of human ether-a-go-go-related gene (HERG) K+ channels expressed in cells HEK-293 and Xenopus oocytes by maprotiline. Maprotiline 177-188 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 16423345-4 2006 Block of HERG expressed in oocytes by maprotiline was enhanced by progressive membrane depolarization and accompanied by a negative shift in the voltage dependence of channel activation. Maprotiline 38-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 16423345-6 2006 The mutation Y652A inverted the voltage dependence of HERG channel block by maprotiline. Maprotiline 76-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 16611127-9 2006 It has been proposed that phenytoin affects the embryonic heart by inhibition of the human-ether-a-go-go (HERG) potassium channel. Phenytoin 26-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-104 16611127-9 2006 It has been proposed that phenytoin affects the embryonic heart by inhibition of the human-ether-a-go-go (HERG) potassium channel. Phenytoin 26-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 16424781-0 2006 Block of HERG channels by berberine: mechanisms of voltage- and state-dependence probed with site-directed mutant channels. Berberine 26-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 16423345-0 2006 Inhibition of cardiac HERG potassium channels by antidepressant maprotiline. Maprotiline 64-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 16288909-0 2006 Clemastine, a conventional antihistamine, is a high potency inhibitor of the HERG K+ channel. Clemastine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 16288909-2 2006 This study investigated the effects on HERG channel current (IHERG) of clemastine, a "conventional" antihistamine that has been associated with delayed ventricular repolarization in vitro, but for which no adverse effects on the human QT interval have been reported. Clemastine 71-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 16288909-8 2006 We conclude that clemastine is a high potency inhibitor of IHERG, that this action is contingent upon channel gating and that clemastine interacts with a high affinity drug-binding site in the HERG channel pore cavity. Clemastine 17-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 16288909-9 2006 The disparity between clemastine"s potent IHERG inhibition and a lack of QT-prolongation in normal clinical use underscores the need to interpret HERG IC50 data for novel compounds in the context of information from other safety assays. Clemastine 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 16472284-4 2006 DNA samples were genotyped for the nucleotide 2690 A>C variation of the KCNH2 gene, corresponding to the KCNH2 K(lysine)897T(threonine) amino acid polymorphism. Lysine 116-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-113 16472284-4 2006 DNA samples were genotyped for the nucleotide 2690 A>C variation of the KCNH2 gene, corresponding to the KCNH2 K(lysine)897T(threonine) amino acid polymorphism. Threonine 128-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-80 16472284-4 2006 DNA samples were genotyped for the nucleotide 2690 A>C variation of the KCNH2 gene, corresponding to the KCNH2 K(lysine)897T(threonine) amino acid polymorphism. Threonine 128-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-113 16219910-10 2006 The effect could be reverted by application of the specific HERG channel inhibitor 4"-[[1-[2-(6-methyl-2-pyridyl)ethyl]-4-piperidinyl]carbonyl]-methanesulfonanilide (E-4031) at 100 nM. 4"-[[1-[2-(6-methyl-2-pyridyl)ethyl]-4-piperidinyl]carbonyl]-methanesulfonanilide 83-164 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 16219910-10 2006 The effect could be reverted by application of the specific HERG channel inhibitor 4"-[[1-[2-(6-methyl-2-pyridyl)ethyl]-4-piperidinyl]carbonyl]-methanesulfonanilide (E-4031) at 100 nM. E 4031 166-172 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 16219910-13 2006 In conclusion, HERG channel activation by small molecules such as NS1643 increases the repolarization reserve and presents an interesting new antiarrhythmic approach. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 66-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 16226079-11 2005 Furthermore, in patients with a mutation in HERG (SQT1), quinidine rendered ventricular tachyarrhythmias noninducible and restored the QT interval/heart rate relationship toward a reference range. Quinidine 57-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15987770-0 2005 {Omega}-3 and {omega}-6 polyunsaturated fatty acids block HERG channels. {omega}-3 and {omega}-6 polyunsaturated fatty acids 0-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 15987770-3 2005 In this study we have analyzed the effects of arachidonic acid (AA, omega-6) and docosahexaenoic acid (DHA, omega-3) on HERG channels stably expressed in Chinese hamster ovary cells by using the whole cell patch-clamp technique. Arachidonic Acid 46-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 15987770-3 2005 In this study we have analyzed the effects of arachidonic acid (AA, omega-6) and docosahexaenoic acid (DHA, omega-3) on HERG channels stably expressed in Chinese hamster ovary cells by using the whole cell patch-clamp technique. Docosahexaenoic Acids 81-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 15987770-3 2005 In this study we have analyzed the effects of arachidonic acid (AA, omega-6) and docosahexaenoic acid (DHA, omega-3) on HERG channels stably expressed in Chinese hamster ovary cells by using the whole cell patch-clamp technique. Docosahexaenoic Acids 103-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 15987770-3 2005 In this study we have analyzed the effects of arachidonic acid (AA, omega-6) and docosahexaenoic acid (DHA, omega-3) on HERG channels stably expressed in Chinese hamster ovary cells by using the whole cell patch-clamp technique. omega-3 108-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 15987770-4 2005 At 10 microM, AA and DHA blocked HERG channels, at the end of 5-s pulses to -10 mV, to a similar extent (37.7 +/- 2.4% vs. 50.2 +/- 8.1%, n = 7-10, P > 0.05). Docosahexaenoic Acids 21-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 15987770-10 2005 Finally, both AA and DHA induced a use-dependent inhibition of HERG channels. Docosahexaenoic Acids 21-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 16309205-3 2005 Microarray analysis and confocal microscopy indicated that there was overexpression of the ether-a-gogo gene (HERG) and the inwardly rectifying potassium channel gene (TWIK) in a human epidermal KB and human liver BEL-7404 carcinoma cell line series selected for cisplatin resistance. Cisplatin 263-272 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 16309205-5 2005 For these reasons, we conducted cell proliferation studies in the presence of either antibodies directed against the detected K+ channels, omeprazole (a H+ pump inhibitor) or a specific inhibitor of the HERG channel (WAY-123398-A-5). way-123398-a 217-229 potassium voltage-gated channel subfamily H member 2 Homo sapiens 203-207 16120615-7 2005 Chromosomal integration of ERG1 ERG7 at their loci in erg26-1ts ets1-1 and erg26-1ts and ets2-1 mutants, respectively, results in the loss of accumulation of squalene and squalene epoxide, re-accumulation of 4-carboxysterols and cell inviability at high temperature. Squalene 158-166 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 16120615-7 2005 Chromosomal integration of ERG1 ERG7 at their loci in erg26-1ts ets1-1 and erg26-1ts and ets2-1 mutants, respectively, results in the loss of accumulation of squalene and squalene epoxide, re-accumulation of 4-carboxysterols and cell inviability at high temperature. oxidosqualene 171-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 16120615-7 2005 Chromosomal integration of ERG1 ERG7 at their loci in erg26-1ts ets1-1 and erg26-1ts and ets2-1 mutants, respectively, results in the loss of accumulation of squalene and squalene epoxide, re-accumulation of 4-carboxysterols and cell inviability at high temperature. 4-carboxysterols 208-224 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 16120615-10 2005 We have mapped mutations within the erg1-1/ets1-1 (G247D) and erg7-1/ets2-1 (D530N, V615E) alleles that suppress the inviability of erg26-1ts at high temperature, and cause accumulation of sterol intermediates and decreased enzymatic activities. Sterols 189-195 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-42 16120615-11 2005 Finally using erg1-1 and erg7-1 mutant strains, we demonstrate that the expression of the ERG25/26/27 genes required for zymosterol biosynthesis are coordinately transcriptionally regulated, along with ERG1 and ERG7, in response to blocks in sterol biosynthesis. zymosterol 121-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-20 16120615-11 2005 Finally using erg1-1 and erg7-1 mutant strains, we demonstrate that the expression of the ERG25/26/27 genes required for zymosterol biosynthesis are coordinately transcriptionally regulated, along with ERG1 and ERG7, in response to blocks in sterol biosynthesis. zymosterol 121-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 202-206 16120615-11 2005 Finally using erg1-1 and erg7-1 mutant strains, we demonstrate that the expression of the ERG25/26/27 genes required for zymosterol biosynthesis are coordinately transcriptionally regulated, along with ERG1 and ERG7, in response to blocks in sterol biosynthesis. Sterols 125-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 202-206 16220205-1 2005 We studied the effects of Chinese traditional medicine rhynchophylline (Rhy) on human ether-a-go-go related gene (HERG) channel and characterized the electrophysiological properties of Rhy"s pharmacological effect on HERG channel using Xenopus oocytes. rhyncophylline 55-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 16166152-0 2005 Tryptophan scanning mutagenesis of the HERG K+ channel: the S4 domain is loosely packed and likely to be lipid exposed. Tryptophan 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 16263765-0 2005 Downregulation of the HERG (KCNH2) K(+) channel by ceramide: evidence for ubiquitin-mediated lysosomal degradation. Ceramides 51-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 16263765-0 2005 Downregulation of the HERG (KCNH2) K(+) channel by ceramide: evidence for ubiquitin-mediated lysosomal degradation. Ceramides 51-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-33 16263765-5 2005 Here we show, using stable HERG-expressing HEK 293 cells, that ceramide evokes a time-dependent decrease in HERG current which was not attributable to a change in gating properties of the channel. Ceramides 63-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 16263765-5 2005 Here we show, using stable HERG-expressing HEK 293 cells, that ceramide evokes a time-dependent decrease in HERG current which was not attributable to a change in gating properties of the channel. Ceramides 63-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 16263765-6 2005 Surface expression of the HERG channel protein was reduced by ceramide as shown by biotinylation of surface proteins, western blotting and immunocytochemistry. Ceramides 62-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 16263765-7 2005 The rapid decline in HERG protein after ceramide stimulation was due to protein ubiquitylation and its association with lysosomes. Ceramides 40-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 16263765-8 2005 The results demonstrate that the surface expression of HERG is strictly regulated, and that ceramide modifies HERG currents and targets the protein for lysosomal degradation. Ceramides 92-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 16260841-0 2005 Coupled K+-water flux through the HERG potassium channel measured by an osmotic pulse method. Water 11-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 16226079-11 2005 Furthermore, in patients with a mutation in HERG (SQT1), quinidine rendered ventricular tachyarrhythmias noninducible and restored the QT interval/heart rate relationship toward a reference range. Quinidine 57-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 16265869-4 2005 The final result of sequencing showed a missense mutation, affecting codon 568 in exon 7 of the KCNH2 (HERG) gene, leading to a tryptophan-cysteine change in the amino acid chain. Tryptophan 128-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-101 16265869-4 2005 The final result of sequencing showed a missense mutation, affecting codon 568 in exon 7 of the KCNH2 (HERG) gene, leading to a tryptophan-cysteine change in the amino acid chain. Tryptophan 128-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 16265869-4 2005 The final result of sequencing showed a missense mutation, affecting codon 568 in exon 7 of the KCNH2 (HERG) gene, leading to a tryptophan-cysteine change in the amino acid chain. Cysteine 139-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-101 16265869-4 2005 The final result of sequencing showed a missense mutation, affecting codon 568 in exon 7 of the KCNH2 (HERG) gene, leading to a tryptophan-cysteine change in the amino acid chain. Cysteine 139-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 15967876-0 2005 Intracellular K+ is required for the inactivation-induced high-affinity binding of cisapride to HERG channels. Cisapride 83-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-100 15983462-1 2005 BACKGROUND: Human ether-a-go-go-related gene (HERG) potassium channels constitute a potential target involved in cardiotoxic side effects of amino-amide local anesthetics. hydrazinide 141-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 15983462-3 2005 The aim of this study was to determine the effect of the mutations Y652A and F656A in the putative drug binding region of HERG on the inhibition by bupivacaine, ropivacaine, and mepivacaine. Bupivacaine 148-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 16155403-1 2005 The antiarrhythmic clofilium is an efficient blocker of hERG1 potassium channels that are strongly expressed in the heart. clofilium 19-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-61 16155403-3 2005 Fluoro- clofilium (F-clofilium) was synthesized and its channel-blocking properties were determined for hERG1 and hEAG1 channels expressed in HEK?293 cells and in Xenopus oocytes. clofilium 0-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-109 16155403-3 2005 Fluoro- clofilium (F-clofilium) was synthesized and its channel-blocking properties were determined for hERG1 and hEAG1 channels expressed in HEK?293 cells and in Xenopus oocytes. clofilium 19-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-109 16155403-6 2005 Similar results were obtained for hERG1, showing F-clofilium is a potent hERG1 and hEAG1 channel blocker once it has reached the intracellularly accessible target site at the channel. clofilium 49-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-39 16155403-6 2005 Similar results were obtained for hERG1, showing F-clofilium is a potent hERG1 and hEAG1 channel blocker once it has reached the intracellularly accessible target site at the channel. clofilium 49-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-78 16011830-1 2005 One variant of the syndrome is linked to missense mutations that lead to a single amino-acid change (N588K; asparagine to lysine) in the S5-Pore linker region of the cardiac HERG K(+) channel. Asparagine 108-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 16011830-1 2005 One variant of the syndrome is linked to missense mutations that lead to a single amino-acid change (N588K; asparagine to lysine) in the S5-Pore linker region of the cardiac HERG K(+) channel. Lysine 122-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 16086867-0 2005 [Correlation of HERG K+ channel protein expression to chemosensitivity of tumor cells to doxorubicin and its modulation by erythromycin]. Doxorubicin 89-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 16086867-0 2005 [Correlation of HERG K+ channel protein expression to chemosensitivity of tumor cells to doxorubicin and its modulation by erythromycin]. Erythromycin 123-135 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 16086867-1 2005 BACKGROUND & OBJECTIVE: Previous studies have found that HERG, a kind of K(+) channel protein, is expressed in some cancers, but its expression is weak or lost in normal tissues. Adenosine Monophosphate 12-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-65 16086867-2 2005 This study was to investigate HERG expression in various cancer cell lines, its correlation with chemosensitivity of cancer cells to doxorubicin, and its biochemical modulation. Doxorubicin 133-144 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 16086867-8 2005 The 50% inhibitory concentration (IC(50)) of doxorubicin was obviously higher in herg-transfected A549 cells than in parent A549 cells. Doxorubicin 45-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 16086867-11 2005 CONCLUSIONS: HERG expression might negatively correlate with the chemosensitivity of tumor cells to doxorubicin. Doxorubicin 100-111 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 15812674-0 2005 HERG K+ channel expression-related chemosensitivity in cancer cells and its modulation by erythromycin. Erythromycin 90-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 15812674-2 2005 In the study reported here, we investigated HERG expression in various cancer cell lines, its correlation with chemosensitivity to vincristine, paclitaxel, and hydroxy-camptothecin, and its biochemical modulation. Vincristine 131-142 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15812674-2 2005 In the study reported here, we investigated HERG expression in various cancer cell lines, its correlation with chemosensitivity to vincristine, paclitaxel, and hydroxy-camptothecin, and its biochemical modulation. Paclitaxel 144-154 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15812674-2 2005 In the study reported here, we investigated HERG expression in various cancer cell lines, its correlation with chemosensitivity to vincristine, paclitaxel, and hydroxy-camptothecin, and its biochemical modulation. hydroxycamptothecinum 160-180 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. Vincristine 204-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. Vincristine 204-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. Vincristine 204-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. Paclitaxel 217-227 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. hydroxycamptothecinum 233-253 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. hydroxycamptothecinum 233-253 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 15812674-7 2005 RESULTS: HERG expression levels differed widely between various human cancer cell lines and HT-29 cells expressing high levels of HERG were more sensitive than A549 cells expressing low levels of HERG to vincristine, paclitaxel, and hydroxy-camptothecin. hydroxycamptothecinum 233-253 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 15812674-8 2005 In terms of IC50, the chemosensitivities of herg-transfected A549 cells to vincristine, paclitaxel and hydroxy-camptothecin were significantly increased. Vincristine 75-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15812674-8 2005 In terms of IC50, the chemosensitivities of herg-transfected A549 cells to vincristine, paclitaxel and hydroxy-camptothecin were significantly increased. Paclitaxel 88-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15812674-8 2005 In terms of IC50, the chemosensitivities of herg-transfected A549 cells to vincristine, paclitaxel and hydroxy-camptothecin were significantly increased. hydroxycamptothecinum 103-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15812674-10 2005 Erythromycin, a HERG K+ channel blocker, suppressed the growth of various cancer cells and the potency was correlated with HERG expression levels. Erythromycin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 15812674-10 2005 Erythromycin, a HERG K+ channel blocker, suppressed the growth of various cancer cells and the potency was correlated with HERG expression levels. Erythromycin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 123-127 15812674-13 2005 There were synergistic effects between erythromycin and vincristine, paclitaxel, and hydroxy-camptothecin, and chemosensitivity was correlated with HERG expression level. Erythromycin 39-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 15812674-13 2005 There were synergistic effects between erythromycin and vincristine, paclitaxel, and hydroxy-camptothecin, and chemosensitivity was correlated with HERG expression level. Paclitaxel 69-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 15812674-14 2005 CONCLUSIONS: HERG expression levels and chemosensitivity were positively correlated for vincristine, paclitaxel, and hydroxy-camptothecin. Vincristine 88-99 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 15812674-14 2005 CONCLUSIONS: HERG expression levels and chemosensitivity were positively correlated for vincristine, paclitaxel, and hydroxy-camptothecin. Paclitaxel 101-111 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 15812674-14 2005 CONCLUSIONS: HERG expression levels and chemosensitivity were positively correlated for vincristine, paclitaxel, and hydroxy-camptothecin. hydroxycamptothecinum 117-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 16051125-6 2005 Under these conditions, erythromycin rapidly blocked I(hERG) even at 22 degrees C. The F656C mutation in the distal S6 of KCNH2 completely abrogated block of I(hERG) measured at 37 degrees C. CONCLUSION: Progressively greater block of hERG and prolongation of APD by erythromycin was observed at temperatures between 36 and 42 degrees C. Temperature-dependent block of I(hERG) is explained by temperature-dependent access of erythromycin to the intracellular binding site at F656. Erythromycin 24-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-127 16051125-6 2005 Under these conditions, erythromycin rapidly blocked I(hERG) even at 22 degrees C. The F656C mutation in the distal S6 of KCNH2 completely abrogated block of I(hERG) measured at 37 degrees C. CONCLUSION: Progressively greater block of hERG and prolongation of APD by erythromycin was observed at temperatures between 36 and 42 degrees C. Temperature-dependent block of I(hERG) is explained by temperature-dependent access of erythromycin to the intracellular binding site at F656. Erythromycin 267-279 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-127 16051125-6 2005 Under these conditions, erythromycin rapidly blocked I(hERG) even at 22 degrees C. The F656C mutation in the distal S6 of KCNH2 completely abrogated block of I(hERG) measured at 37 degrees C. CONCLUSION: Progressively greater block of hERG and prolongation of APD by erythromycin was observed at temperatures between 36 and 42 degrees C. Temperature-dependent block of I(hERG) is explained by temperature-dependent access of erythromycin to the intracellular binding site at F656. Erythromycin 267-279 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-127 15983462-3 2005 The aim of this study was to determine the effect of the mutations Y652A and F656A in the putative drug binding region of HERG on the inhibition by bupivacaine, ropivacaine, and mepivacaine. Ropivacaine 161-172 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 15983462-3 2005 The aim of this study was to determine the effect of the mutations Y652A and F656A in the putative drug binding region of HERG on the inhibition by bupivacaine, ropivacaine, and mepivacaine. Mepivacaine 178-189 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 15983462-4 2005 METHODS: The authors examined the inhibition of wild-type and mutant HERG channels, transiently expressed in Chinese hamster ovary cells by bupivacaine, ropivacaine, and mepivacaine. Bupivacaine 140-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15983462-4 2005 METHODS: The authors examined the inhibition of wild-type and mutant HERG channels, transiently expressed in Chinese hamster ovary cells by bupivacaine, ropivacaine, and mepivacaine. Ropivacaine 153-164 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15983462-4 2005 METHODS: The authors examined the inhibition of wild-type and mutant HERG channels, transiently expressed in Chinese hamster ovary cells by bupivacaine, ropivacaine, and mepivacaine. Mepivacaine 170-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15983462-10 2005 The mutations resulted in a change of the stereoselectivity of HERG channel block by ropivacaine. Ropivacaine 85-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 15967876-6 2005 In the present study, we examined the effect of inactivation gating on cisapride block of HERG. Cisapride 71-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 15967876-7 2005 Modulation of HERG inactivation was achieved by either changing extracellular K+ or Cs+ concentrations or by mutations of the channel. Cesium 84-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 15967876-9 2005 Furthermore, cisapride block of the HERG K+ current was not linked with inactivation in the mutant HERG channels F656V and F656M. Cisapride 13-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 15967876-10 2005 Our results suggest that inactivation facilitates cisapride block of HERG channels through affecting the positioning of Phe-656. Cisapride 50-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15967876-10 2005 Our results suggest that inactivation facilitates cisapride block of HERG channels through affecting the positioning of Phe-656. Phenylalanine 120-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15961404-1 2005 Mutations of a putative cyclic-nucleotide-binding domain (CNBD) can disrupt the function of the hyperpolarization-activated cyclic-nucleotide-gated channel (HCN2) and the human ether-a-go-go-related gene potassium channel (HERG). Nucleotides, Cyclic 24-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 223-227 16076272-11 2005 Pharmacologic treatment has only been investigated in patients with a mutation in KCNH2 (HERG), and it could be demonstrated that the mutant currents may be insufficiently suppressed by drugs that are targeted to block the specific current (e.g., sotalol or ibutilide) in patients with a mutation in the IK(r-)coding gene KCNH2 (HERG). ibutilide 258-267 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 16041196-4 2005 Since the HERG gene encodes IKr, we studied quinidine"s effect on HERG expressed in Xenopus oocytes by the 2-electrode voltage clamp technique. Quinidine 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 10-14 16041196-4 2005 Since the HERG gene encodes IKr, we studied quinidine"s effect on HERG expressed in Xenopus oocytes by the 2-electrode voltage clamp technique. Quinidine 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 16041196-5 2005 When extracellular K+ was 5 mmol/L, quinidine blocked the HERG current dose dependently, with an IC50 of 6.3 +/- 0.2 micromol/L. Quinidine 36-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 16041196-7 2005 The inhibition of HERG by quinidine was not use dependent. Quinidine 26-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 15842772-0 2005 Modulating effect of ginseng saponins on heterologously expressed HERG currents in Xenopus oocytes. Saponins 29-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 15711590-4 2005 The drug pentamidine, at near therapeutic concentrations that do not cause direct KCNH2 channel block, disrupts normal KCNH2 channel protein processing and maturation to reduce its surface membrane expression. Pentamidine 9-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 119-124 15973763-7 2005 The mutation might affect, through deficient splicing, the putative cyclic nucleotide binding domain (CNBD) of the HERG K(+) channel. Nucleotides, Cyclic 68-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 15800067-8 2005 hERG1-transfected cells undergo an activation of hERG currents after beta(1) integrin-mediated adhesion to fibronectin; concomitant with this activation, the focal adhesion kinase associates with the hERG1 protein and becomes tyrosine phosphorylated. Tyrosine 226-234 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-5 15800067-9 2005 Using hERG1-specific inhibitors, we show that the tyrosine phosphorylation of focal adhesion kinase is strictly dependent on hERG channel activity. Tyrosine 50-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 6-11 16007460-0 2005 Inhibition of cardiac HERG channels by grapefruit flavonoid naringenin: implications for the influence of dietary compounds on cardiac repolarisation. Flavonoids 50-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 16007460-0 2005 Inhibition of cardiac HERG channels by grapefruit flavonoid naringenin: implications for the influence of dietary compounds on cardiac repolarisation. naringenin 60-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 16007460-2 2005 In a previous study, we demonstrated for the first time that flavonoids are inhibitors of cardiac human ether-a-go-go-related gene (HERG) channels. Flavonoids 61-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-136 16007460-4 2005 HERG blockade by grapefruit flavonoid naringenin is most likely to be the mechanism underlying this effect. Flavonoids 28-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 16007460-4 2005 HERG blockade by grapefruit flavonoid naringenin is most likely to be the mechanism underlying this effect. naringenin 38-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 16007460-5 2005 Therefore, the electrophysiological properties of HERG blockade by naringenin were analysed in detail. naringenin 67-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 16007460-6 2005 HERG potassium currents expressed in Xenopus oocytes were measured with a two-microelectrode voltage clamp. Potassium 5-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 16007460-7 2005 Naringenin blocked HERG potassium channels with an IC50 value of 102.6 microM in Xenopus oocytes. naringenin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 16007460-10 2005 Naringenin binding to HERG required aromatic residue F656 in the putative pore binding site. naringenin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 16007460-15 2005 Naringenin inhibits HERG channels with pharmacological characteristics similar to those of well-known HERG antagonists. naringenin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 16007460-15 2005 Naringenin inhibits HERG channels with pharmacological characteristics similar to those of well-known HERG antagonists. naringenin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 15760896-9 2005 These results suggest that the degradation of LQT2 mutant channels is mediated by the cytosolic proteasome in a process that involves mannose trimming, polyubiquitination, and deglycosylation of mutant channels. Mannose 134-141 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 15842772-1 2005 AIM: To examine the effects of ginseng saponins on the heterologously expressed human ether-a-go-go related gene (HERG) that encodes the rapid component of the delayed rectifier K+ channel. Saponins 39-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 15842772-4 2005 RESULTS: Crude saponins of Korean red ginseng (GS) induced a minimal increase of the maximal HERG conductance without changes in the voltage-dependent HERG current activation and inactivation curves. Saponins 15-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15842772-5 2005 GS, however, decelerated HERG current deactivation in a concentration-dependent manner, which was more noticeable with panaxitriol (PT) than panaxidiol (PD). panaxitriol 119-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 15842772-5 2005 GS, however, decelerated HERG current deactivation in a concentration-dependent manner, which was more noticeable with panaxitriol (PT) than panaxidiol (PD). Platinum 132-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 15842772-5 2005 GS, however, decelerated HERG current deactivation in a concentration-dependent manner, which was more noticeable with panaxitriol (PT) than panaxidiol (PD). panaxidiol 141-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 15842772-6 2005 Consistently, ginseng saponins increased the HERG deactivation time constants with the order of potency of Rg1 (a major component of PT)>Rf1>Rb1 (a major component of PD). Saponins 22-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 15842772-9 2005 CONCLUSION: Ginseng saponins enhance HERG currents, which could be in part a possible mechanism of the shortening cardiac action potential of ginseng saponins. Saponins 20-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 15842772-9 2005 CONCLUSION: Ginseng saponins enhance HERG currents, which could be in part a possible mechanism of the shortening cardiac action potential of ginseng saponins. Saponins 150-158 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 15814090-5 2005 Verapamil was used as an HERG channel blocker that is not associated with TdP. Verapamil 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 15722405-2 2005 We studied the effects of trifluoperazine on the human ether-a-go-go-related gene (HERG) channel expressed in Xenopus oocytes and on the delayed rectifier K(+) current of guinea pig cardiomyocytes. Trifluoperazine 26-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 15722405-3 2005 The application of trifluoperazine showed a dose-dependent decrease in current amplitudes at the end of voltage steps and tail currents of HERG. Trifluoperazine 19-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-143 15722405-4 2005 The IC(50) for a trifluoperazine block of HERG current progressively decreased according to depolarization: IC(50) values at -40, 0, and +40 mV were 21.6, 16.6, and 9.29 microM, respectively. Trifluoperazine 17-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 15722405-6 2005 The block of HERG by trifluoperazine was use-dependent, exhibiting more rapid onset and greater steady-state block at higher frequencies of activation; there was partial relief of the block with decreasing frequency. Trifluoperazine 21-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 15640612-0 2005 Comparison of the effects of metoclopramide and domperidone on HERG channels. Metoclopramide 29-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 15722405-8 2005 Our findings suggest the arrhythmogenic side effect of trifluoperazine is caused by a blockade of HERG and the rapid component of the delayed rectifier K(+) current rather than by the blockade of the slow component. Trifluoperazine 55-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 15640612-0 2005 Comparison of the effects of metoclopramide and domperidone on HERG channels. Domperidone 48-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 15640612-3 2005 The gastroprokinetic agent cisapride is a potent blocker of HERG currents and serious cardiac arrhythmias and deaths from TdP and ventricular fibrillation have been reported in patients taking cisapride. Cisapride 27-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 15640612-3 2005 The gastroprokinetic agent cisapride is a potent blocker of HERG currents and serious cardiac arrhythmias and deaths from TdP and ventricular fibrillation have been reported in patients taking cisapride. Cisapride 193-202 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 15640612-4 2005 The aim of the present study was to compare the effects of the gastroprokinetic agents domperidone and metoclopramide on HERG channels transiently expressed in human embryonic kidney (HEK 293) cells using the whole-cell configuration of the patch-clamp technique. Domperidone 87-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 15640612-4 2005 The aim of the present study was to compare the effects of the gastroprokinetic agents domperidone and metoclopramide on HERG channels transiently expressed in human embryonic kidney (HEK 293) cells using the whole-cell configuration of the patch-clamp technique. Metoclopramide 103-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 15640612-5 2005 Both domperidone and metoclopramide concentration-dependently blocked HERG currents, and the following values were calculated for IC(50) (the concentrations causing half-maximal inhibition) and n (the Hill coefficient): 57.0 nmol/l and 0.99 for domperidone, 5.4 micromol/l and 0.95 for metoclopramide. Domperidone 5-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 15640612-5 2005 Both domperidone and metoclopramide concentration-dependently blocked HERG currents, and the following values were calculated for IC(50) (the concentrations causing half-maximal inhibition) and n (the Hill coefficient): 57.0 nmol/l and 0.99 for domperidone, 5.4 micromol/l and 0.95 for metoclopramide. Metoclopramide 21-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Domperidone 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Metoclopramide 154-168 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Metoclopramide 154-168 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Domperidone 241-252 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Domperidone 241-252 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Metoclopramide 257-271 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 15640612-6 2005 The observation that the extent of block of HERG currents by domperidone increased at more positive membrane potentials whereas block of HERG currents by metoclopramide displayed a smaller degree of voltage dependency seems to indicate that domperidone and metoclopramide have distinct binding sites on HERG channels. Metoclopramide 257-271 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 15640612-7 2005 In conclusion, the potency for block of HERG currents is about 100-fold lower for metoclopramide when compared to domperidone. Metoclopramide 82-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 15640612-7 2005 In conclusion, the potency for block of HERG currents is about 100-fold lower for metoclopramide when compared to domperidone. Domperidone 114-125 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 15778703-0 2005 Blockade of HERG cardiac K+ current by antifungal drug miconazole. Miconazole 55-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 15778703-5 2005 To determine the mechanism underlying these clinical findings, we investigated the effect of miconazole on human ether-a-go-go-related gene (HERG) K+ channels. Miconazole 93-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 141-145 15778703-9 2005 Miconazole inhibited HERG peak tail current in a concentration-dependent manner (0.4-40 microM) with an IC50 of 2.1 microM (n=3-5 cells at each concentration, Hill coefficient 1.2). Miconazole 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 15778703-16 2005 The S6 domain mutation, F656C, abolished the inhibitory action of miconazole on HERG current indicating that miconazole preferentially binds to an aromatic amino-acid residue within the pore-S6 region. Miconazole 66-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 15778703-16 2005 The S6 domain mutation, F656C, abolished the inhibitory action of miconazole on HERG current indicating that miconazole preferentially binds to an aromatic amino-acid residue within the pore-S6 region. Miconazole 109-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 15778703-16 2005 The S6 domain mutation, F656C, abolished the inhibitory action of miconazole on HERG current indicating that miconazole preferentially binds to an aromatic amino-acid residue within the pore-S6 region. Amino Acids, Aromatic 147-166 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 15778703-18 2005 Our findings indicate that miconazole causes HERG channel block by binding to a common drug receptor, and this involves preferential binding to activated channels. Miconazole 27-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 15778703-19 2005 Thus, miconazole prolongs the QT interval by direct inhibition of HERG channels. Miconazole 6-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 16244363-6 2005 Consistent with ATF6 activation, the ER chaperones/calcium-binding proteins Grp78, Grp94, and calreticulin are elevated in I593R HERG-expressing cells. Calcium 51-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 129-133 15846098-0 2005 The potent inhibitory effects of cisapride, a specific blocker for human ether-a-go-go-related gene (HERG) channel, on gastric cancer cells. Cisapride 33-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 15846098-2 2005 Human ether-a-go-go-related gene (HERG) encoding one of the components of delayed rectifier potassium currents has been indicated to be involved in tumor cell growth and death. Potassium 92-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 15846098-3 2005 Our aim is to investigate the effects of cisapride, a specific blocker for HERG channel, on human gastric cancer cells. Cisapride 41-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-79 15846098-8 2005 The proliferation of gastric cancer cells expressing HERG protein was inhibited in a time- and dose-dependent manner when treated with cisapride (P<0.05). Cisapride 135-144 potassium voltage-gated channel subfamily H member 2 Homo sapiens 53-57 15846098-12 2005 CONCLUSIONS: As HERG channel blocker, cisapride, can inhibit the growth of gastric cancer cells by altering distribution of cell cycle and inducing apoptosis so as to be of potential value in the treatment of gastric cancer. Cisapride 38-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 15821840-5 2005 RESULTS: Quinidine (10 microM) inhibited HERG tail current by 37% +/- 5% at pH7.4. Quinidine 9-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 15821840-7 2005 Dofetilide (0.3 microM) inhibited HERG tail current by 34% +/- 3% and 1% +/- 2% at extracellular pH 7.4 and 6.2, respectively. dofetilide 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Lopinavir 138-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Lopinavir 138-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 218-222 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Nelfinavir 149-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Nelfinavir 149-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 218-222 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Ritonavir 161-170 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Ritonavir 161-170 potassium voltage-gated channel subfamily H member 2 Homo sapiens 218-222 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Saquinavir 176-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15721475-5 2005 Drug-induced QT prolongation is usually caused by block of human ether-a-go-go-related gene (HERG) potassium channels, and we showed that lopinavir, nelfinavir, ritonavir, and saquinavir caused dose-dependent block of HERG channels heterologously expressed in HEK293 cells in vitro. Saquinavir 176-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 218-222 15821840-8 2005 Azimilide (10 microM) inhibited HERG tail current by 59% +/- 3% and 17% +/- 3% at extracellular pH 7.4 and 6.2. azimilide 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 15821840-9 2005 There were significant differences in the HERG inhibition by quinidine, dofetilide, and azimilide between pH 7.4 and pH 6.2 (P < .01). Quinidine 61-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 15821840-9 2005 There were significant differences in the HERG inhibition by quinidine, dofetilide, and azimilide between pH 7.4 and pH 6.2 (P < .01). dofetilide 72-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 15821840-9 2005 There were significant differences in the HERG inhibition by quinidine, dofetilide, and azimilide between pH 7.4 and pH 6.2 (P < .01). azimilide 88-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 15548764-7 2005 RPR260243-modified HERG currents were inhibited by dofetilide (IC50 = 58 nM). dofetilide 51-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 15733182-1 2005 AIMS: To explore effects of epinephrine and phenylephrine on the behavior of right ventricular monophasic action potentials (MAPs) in symptomatic LQT1 and LQT2 patients. Epinephrine 28-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 15733182-1 2005 AIMS: To explore effects of epinephrine and phenylephrine on the behavior of right ventricular monophasic action potentials (MAPs) in symptomatic LQT1 and LQT2 patients. Phenylephrine 44-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 15679475-4 2005 Propranolol caused a concentration-dependent inhibition of HERG current with an IC50 value of 81 microM at -10 mV. Propranolol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 15679475-7 2005 The propranolol analogue ICI118551 ((+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride) blocked the HERG channel with similar affinity, whereas the beta1-receptor antagonists metoprolol and atenolol showed weak effects. Propranolol 4-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 15679475-7 2005 The propranolol analogue ICI118551 ((+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride) blocked the HERG channel with similar affinity, whereas the beta1-receptor antagonists metoprolol and atenolol showed weak effects. ICI 118551 25-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 15679475-7 2005 The propranolol analogue ICI118551 ((+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride) blocked the HERG channel with similar affinity, whereas the beta1-receptor antagonists metoprolol and atenolol showed weak effects. ((+/-)-1-[2,3-(dihydro-7-methyl-1h-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride 35-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 15500450-0 2005 Compound heterozygosity for mutations Asp611-->Tyr in KCNQ1 and Asp609-->Gly in KCNH2 associated with severe long QT syndrome. Glycine 79-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-91 15500450-2 2005 We have identified two heterozygous missense mutations in the KCNQ1 and KCNH2 (also known as HERG) genes [Asp611-->Tyr (D611Y) in KCNQ1 and Asp609-->Gly (D609G) in KCNH2] in a 2-year-old boy with LQTS. Tyrosine 118-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 15500450-2 2005 We have identified two heterozygous missense mutations in the KCNQ1 and KCNH2 (also known as HERG) genes [Asp611-->Tyr (D611Y) in KCNQ1 and Asp609-->Gly (D609G) in KCNH2] in a 2-year-old boy with LQTS. Tyrosine 118-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15500450-2 2005 We have identified two heterozygous missense mutations in the KCNQ1 and KCNH2 (also known as HERG) genes [Asp611-->Tyr (D611Y) in KCNQ1 and Asp609-->Gly (D609G) in KCNH2] in a 2-year-old boy with LQTS. Tyrosine 118-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 170-175 15500450-2 2005 We have identified two heterozygous missense mutations in the KCNQ1 and KCNH2 (also known as HERG) genes [Asp611-->Tyr (D611Y) in KCNQ1 and Asp609-->Gly (D609G) in KCNH2] in a 2-year-old boy with LQTS. Glycine 155-158 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 15500450-2 2005 We have identified two heterozygous missense mutations in the KCNQ1 and KCNH2 (also known as HERG) genes [Asp611-->Tyr (D611Y) in KCNQ1 and Asp609-->Gly (D609G) in KCNH2] in a 2-year-old boy with LQTS. Glycine 155-158 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 15851286-8 2005 During epinephrine infusion, G1- and G2-T waves were more common in LQT2 than in LQT1 (75% vs 26%, P = .009). Epinephrine 7-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 15851286-10 2005 Epinephrine-precipitated biphasic T waves were observed similarly in all groups: LQT1 (6/30), LQT2 (3/28), and control (4/32). Epinephrine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 15851286-11 2005 During low-dose epinephrine infusion (< or =0.05 microg/kg/min), G1-T waves occurred more frequently in LQT2 (LQT1: 25% vs 3%; control 9%, P = .02). Epinephrine 16-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-111 15851286-12 2005 Low-dose epinephrine-induced G2-T waves were detected exclusively in LQT2 (18%). Epinephrine 9-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15851286-16 2005 G2-notched T waves elicited during low-dose epinephrine may unmask some patients with concealed LQT2. Epinephrine 44-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-100 15608471-6 2005 This data, associated with the finding that methadone is a rather potent inhibitor of HERG potassium channels and that it may induce torsade de pointes in predisposed subjects, supports the recommendation that patients entering methadone treatment (MT) are screened for cardiac risk factors. Methadone 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 15670565-1 2005 In a 7-week-old infant who experienced sudden infant death syndrome (SIDS), a novel missense mutation was identified in KCNH2, causing a lysine-to-glutamic acid amino acid substitution at position 101 (K101E). Lysine 137-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-125 15670565-1 2005 In a 7-week-old infant who experienced sudden infant death syndrome (SIDS), a novel missense mutation was identified in KCNH2, causing a lysine-to-glutamic acid amino acid substitution at position 101 (K101E). glutamic acid amino acid 147-171 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-125 15673388-0 2005 Further insights into the effect of quinidine in short QT syndrome caused by a mutation in HERG. Quinidine 36-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 15673388-1 2005 INTRODUCTION: The principal aim of this study was to assess the efficacy of quinidine in suppressing IKr in vitro and in modulating the rate dependence of the QT interval in the "SQT1" form of the short QT syndrome. Quinidine 76-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 15673388-6 2005 Data from heterologous expression of wild-type and mutant HERG genes indicate the mutation causes a 20-fold increase in IC50 of d-sotalol but only a 5.8-fold increase in IC50 of quinidine. Dexsotalol 128-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 15673388-6 2005 Data from heterologous expression of wild-type and mutant HERG genes indicate the mutation causes a 20-fold increase in IC50 of d-sotalol but only a 5.8-fold increase in IC50 of quinidine. Quinidine 178-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 15673388-7 2005 CONCLUSION: Oral quinidine is effective in suppressing the gain of function in IKr responsible for some cases of short QT syndrome with a mutation in HERG and thus restoring normal rate dependence of the QT interval and rendering ventricular tachycardia/ventricular fibrillation noninducible. Quinidine 17-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 150-154 15572053-7 2004 Using sucrose gradient centrifugation we showed that wild type HERG and R1014X formed a tetrameric structure, whereas Q725X was expressed as a monomer. Sucrose 6-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 15541373-0 2004 High affinity HERG K(+) channel blockade by the antiarrhythmic agent dronedarone: resistance to mutations of the S6 residues Y652 and F656. Dronedarone 69-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 15541373-2 2004 Two aromatic amino-acid residues (Y652 and F656) on the inner (S6) helices are considered to be key constituents of a high affinity drug binding site within the HERG channel pore cavity. Amino Acids, Aromatic 4-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 161-165 15541373-3 2004 Using wild-type (WT) and mutant HERG channels expressed in mammalian cell lines, we have investigated HERG channel current (I(HERG)) blockade at 37+/-1 degrees C by dronedarone (DRONED), a non-iodinated analogue of the Class III antiarrhythmic agent amiodarone (AMIOD). Dronedarone 165-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 15541373-3 2004 Using wild-type (WT) and mutant HERG channels expressed in mammalian cell lines, we have investigated HERG channel current (I(HERG)) blockade at 37+/-1 degrees C by dronedarone (DRONED), a non-iodinated analogue of the Class III antiarrhythmic agent amiodarone (AMIOD). Dronedarone 165-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 15558241-0 2004 Inhibitory effects of AMP 579, a novel cardioprotective adenosine A1/A2A receptor agonist, on native IKr and cloned HERG current. 4-(7-((2-3-chloro-2-thienyl)-1-methyl-propylamino)-3H-imidazo(4,5-b)pyridyl-3-yl)cyclopentane carboxamide 22-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 116-120 15599107-1 2004 While aldosterone receptor blockers improve survival of patients with congestive heart failure, spironolactone and its derivatives were recently shown to block ether-a-go-go-related gene (HERG) channels and native IKs and IKr currents in guinea pig ventricular myocytes. Spironolactone 96-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 188-192 15558241-7 2004 In HEK 293 cells expressing HERG channels, AMP 579 (10 microM) significantly blocked the HERG current at +10 mV by 34.9+/-7.0% (n=4, p<0.05), and the degree of inhibition was comparable with that observed in guinea-pig ventricular myocytes (36.8+/-6.0%, n=4). Adenosine Monophosphate 43-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-32 15558241-7 2004 In HEK 293 cells expressing HERG channels, AMP 579 (10 microM) significantly blocked the HERG current at +10 mV by 34.9+/-7.0% (n=4, p<0.05), and the degree of inhibition was comparable with that observed in guinea-pig ventricular myocytes (36.8+/-6.0%, n=4). Adenosine Monophosphate 43-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 15558241-10 2004 Thus, AMP 579 inhibits both native I(Kr) and cloned HERG channels with additional inhibitory effect of I(Ca), and such inhibitory effects may at least partially underlie the observed antifibrillatory action of the drug during myocardial ischemia/reperfusion. Adenosine Monophosphate 6-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 15558243-4 2004 Here we show that halofantrine preferentially blocks open and inactivated HERG channels heterologously expressed in Xenopus laevis oocytes. halofantrine 18-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 15599706-0 2004 Class Ia anti-arrhythmic drug ajmaline blocks HERG potassium channels: mode of action. Ajmaline 30-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 15558243-6 2004 As we reported previously for other HERG channel blockers, the potency of halofantrine was reduced by mutation to Ala of aromatic residues (Y652, F656) located in the S6 domain, or a Val (V625) located in the pore helix. halofantrine 74-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 15599706-5 2004 Ajmaline blocked HERG currents with an IC(50) of 1.0 micromol/l in HEK cells and 42.3 micromol/l in Xenopus oocytes. Ajmaline 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 15599706-7 2004 In HERG mutant channels Y652A and F656A lacking aromatic residues in the S6 domain, the inhibitory effect of ajmaline was completely abolished. Ajmaline 109-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 15558243-6 2004 As we reported previously for other HERG channel blockers, the potency of halofantrine was reduced by mutation to Ala of aromatic residues (Y652, F656) located in the S6 domain, or a Val (V625) located in the pore helix. Alanine 114-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 15599706-8 2004 Ajmaline induced a small shift in HERG current half-maximal activation voltage towards more negative potentials. Ajmaline 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 15558243-12 2004 We conclude that halofantrine requires channels to open before it can gain access to its binding site located in the central cavity of the HERG channel. halofantrine 17-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-143 15599706-12 2004 Ajmaline blocked HERG channels in the open, but not in the closed states. Ajmaline 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 15599706-13 2004 Binding of ajmaline to inactivated HERG channels may also be possible. Ajmaline 11-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 15599706-3 2004 In order to elucidate the molecular basis of these effects, we examined effects of ajmaline on human ether a-go-go related gene HERG potassium channels. Ajmaline 83-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 15599706-14 2004 In inactivation-deficient HERG S620T channels, the sensitivity to ajmaline was markedly reduced. Ajmaline 66-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 15599706-15 2004 The IC(50) of HERG channel blockade in HEK cells lies within the range of unbound therapeutic plasma concentrations of ajmaline. Ajmaline 119-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 15599706-16 2004 Therefore, inhibitory effects on HERG channels may contribute to both the high anti-arrhythmic efficacy of ajmaline and to its pro-arrhythmic potential. Ajmaline 107-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 15322737-0 2004 Inhibition of cardiac HERG potassium channels by the atypical antidepressant trazodone. Trazodone 77-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 15280442-4 2004 Using the patch-clamp technique, we showed that clobutinol dose-dependently inhibited the HERG K(+) current with a half-maximum block concentration of 2.9 microM. clobutinol 48-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 15851169-2 2004 BACKGROUND: A differential response of dynamic QT interval to epinephrine infusion between LQT1, LQT2, and LQT3 syndromes has been reported, indicating the potential diagnostic value of the epinephrine test for genotyping the three forms. Epinephrine 62-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 15851169-2 2004 BACKGROUND: A differential response of dynamic QT interval to epinephrine infusion between LQT1, LQT2, and LQT3 syndromes has been reported, indicating the potential diagnostic value of the epinephrine test for genotyping the three forms. Epinephrine 190-201 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 15851169-6 2004 RESULTS: The sensitivity (penetrance) by ECG diagnostic criteria was lower in LQT1 (68%) than in LQT2 (83%) or LQT3 (83%) before epinephrine and was improved with steady-state epinephrine in LQT1 (87%) and LQT2 (91%) but not in LQT3 (83%), without the expense of specificity (100%). Epinephrine 176-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 206-210 15851169-9 2004 CONCLUSIONS: Epinephrine infusion is a powerful test to predict the genotype of LQT1, LQT2, and LQT3 syndromes as well as to improve the clinical diagnosis of genotype-positive patients, especially those with LQT1 syndrome. Epinephrine 13-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 15280551-7 2004 This clinical result is supported by in vitro studies of HERG dofetilide sensitivity by using coexpression of HERG with wild-type and I447V KCR1 cDNAs. dofetilide 62-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 15280551-7 2004 This clinical result is supported by in vitro studies of HERG dofetilide sensitivity by using coexpression of HERG with wild-type and I447V KCR1 cDNAs. dofetilide 62-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 15072950-2 2004 We previously showed that the HERG N470D mutation expressed as homotetrameric channels causes a protein trafficking defect, and this can be corrected by the HERG channel blocking drug E-4031. E 4031 184-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 15072950-2 2004 We previously showed that the HERG N470D mutation expressed as homotetrameric channels causes a protein trafficking defect, and this can be corrected by the HERG channel blocking drug E-4031. E 4031 184-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 157-161 15685300-4 2005 Changing from supine to standing alone caused a significant increase in QTc in the LQT2 group compared with in control subjects (change in QTc of 48+/-38 ms versus 21+/-29 ms, respectively, P=0.02). qtc 72-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 15231497-0 2004 Molecular analysis of PIP2 regulation of HERG and IKr. Phosphatidylinositol 4,5-Diphosphate 22-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 15231497-1 2004 We previously reported that cloned human ether-a-go-go-related gene (HERG) K+ channels are regulated by changes in phosphatidylinositol 4,5-bisphosphate (PIP2) concentration. Phosphatidylinositol 4,5-Diphosphate 115-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15231497-1 2004 We previously reported that cloned human ether-a-go-go-related gene (HERG) K+ channels are regulated by changes in phosphatidylinositol 4,5-bisphosphate (PIP2) concentration. Phosphatidylinositol 4,5-Diphosphate 154-158 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 15231497-2 2004 Here we investigated the molecular determinants of PIP2 interactions with HERG channel protein. Phosphatidylinositol 4,5-Diphosphate 51-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 15231497-6 2004 Elevating internal PIP2, however, no longer accelerated the activation kinetics of the mutant HERG. Phosphatidylinositol 4,5-Diphosphate 19-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 15231497-12 2004 These results support a physiological role for PIP2 regulation of the rapidly activating delayed rectifier K+ current during autonomic stimulation and localize a site of interaction to the COOH-terminal tail of the HERG K+ channel. Phosphatidylinositol 4,5-Diphosphate 47-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 215-219 15522280-0 2004 Role of a KCNH2 polymorphism (R1047 L) in dofetilide-induced Torsades de Pointes. dofetilide 42-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 10-15 15308760-0 2004 The low-potency, voltage-dependent HERG blocker propafenone--molecular determinants and drug trapping. Propafenone 48-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 15308760-1 2004 The molecular determinants of high-affinity human ether-a-go-go-related gene (HERG) potassium channel blockade by methanesulfonanilides include two aromatic residues (Phe656 and Tyr652) on the inner helices (S6) and residues on the pore helices that face into the inner cavity, but determinants for lower-affinity HERG blockers may be different. methanesulfonanilides 114-135 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 15308760-1 2004 The molecular determinants of high-affinity human ether-a-go-go-related gene (HERG) potassium channel blockade by methanesulfonanilides include two aromatic residues (Phe656 and Tyr652) on the inner helices (S6) and residues on the pore helices that face into the inner cavity, but determinants for lower-affinity HERG blockers may be different. methanesulfonanilides 114-135 potassium voltage-gated channel subfamily H member 2 Homo sapiens 314-318 15308760-2 2004 In this study, alanine-substituted HERG channel mutants of inner cavity residues were expressed in Xenopus laevis oocytes and were used to characterize the HERG channel binding site of the antiarrhythmic propafenone. Alanine 15-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 15308760-2 2004 In this study, alanine-substituted HERG channel mutants of inner cavity residues were expressed in Xenopus laevis oocytes and were used to characterize the HERG channel binding site of the antiarrhythmic propafenone. Alanine 15-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 156-160 15308760-2 2004 In this study, alanine-substituted HERG channel mutants of inner cavity residues were expressed in Xenopus laevis oocytes and were used to characterize the HERG channel binding site of the antiarrhythmic propafenone. Propafenone 204-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 15308760-2 2004 In this study, alanine-substituted HERG channel mutants of inner cavity residues were expressed in Xenopus laevis oocytes and were used to characterize the HERG channel binding site of the antiarrhythmic propafenone. Propafenone 204-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 156-160 15308760-3 2004 Propafenone"s blockade of HERG was strongly dependent on residue Phe656 but was insensitive or weakly sensitive to mutation of Tyr652, Thr623, Ser624, Val625, Gly648, or Val659 and did not require functional inactivation. Propafenone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 15308760-4 2004 Homology models of HERG based on KcsA and MthK crystal structures, representing the closed and open forms of the channel, respectively, suggest propafenone is trapped in the inner cavity and is unable to interact exclusively with Phe656 in the closed state (whereas exclusive interactions between propafenone and Phe656 are found in the open-channel model). Propafenone 144-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 15476742-4 2004 We studied the ability of three compounds that inhibit PDE5--sildenafil, tadalafil, and vardenafil--to block the HERG channel. Sildenafil Citrate 61-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 15476742-4 2004 We studied the ability of three compounds that inhibit PDE5--sildenafil, tadalafil, and vardenafil--to block the HERG channel. Tadalafil 73-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 15476742-4 2004 We studied the ability of three compounds that inhibit PDE5--sildenafil, tadalafil, and vardenafil--to block the HERG channel. Vardenafil Dihydrochloride 88-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 15476742-8 2004 Because the maximum soluble concentration of tadalafil (100 microM) produced only a 50.9% inhibition of the HERG current amplitude, the IC50 value for tadalafil could not be determined with the Hill equation. Tadalafil 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 15476742-9 2004 Tadalafil had the weakest capacity to block the HERG channel, producing a 50.9% blockade at the maximum soluble concentration (100 microM), compared with 86.2% for vardenafil (100 microM) and 75.2% for sildenafil (100 microM). Tadalafil 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 15476742-9 2004 Tadalafil had the weakest capacity to block the HERG channel, producing a 50.9% blockade at the maximum soluble concentration (100 microM), compared with 86.2% for vardenafil (100 microM) and 75.2% for sildenafil (100 microM). Vardenafil Dihydrochloride 164-174 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 15476742-9 2004 Tadalafil had the weakest capacity to block the HERG channel, producing a 50.9% blockade at the maximum soluble concentration (100 microM), compared with 86.2% for vardenafil (100 microM) and 75.2% for sildenafil (100 microM). Sildenafil Citrate 202-212 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 15169846-0 2004 Functional interaction between extracellular sodium, potassium and inactivation gating in HERG channels. Sodium 45-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 15169846-0 2004 Functional interaction between extracellular sodium, potassium and inactivation gating in HERG channels. Potassium 53-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 15266014-0 2004 Structural determinants of HERG channel block by clofilium and ibutilide. clofilium 49-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 15266014-0 2004 Structural determinants of HERG channel block by clofilium and ibutilide. ibutilide 63-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 15266014-7 2004 Clofilium, a chlorobenzene derivative, is a potent blocker of HERG channels, but has a remarkably slower time course for recovery from block than ibutilide, a methanesulfonanilide. clofilium 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 15266014-9 2004 There is little recovery from clofilium block with D540K HERG channels that permit untrapping at hyperpolarized potentials. clofilium 30-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 15266014-11 2004 However, S624A, located at the base of the pore helix, was the only HERG mutation that enabled rapid recovery from clofilium block. clofilium 115-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 15266014-12 2004 In summary, the pore helix residues are important components of the HERG drug binding site, and may be particularly important for drugs with polar substituents, such as a halogen (e.g., clofilium) or a methanesulfonamide (e.g., ibutilide). Halogens 171-178 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 15266014-12 2004 In summary, the pore helix residues are important components of the HERG drug binding site, and may be particularly important for drugs with polar substituents, such as a halogen (e.g., clofilium) or a methanesulfonamide (e.g., ibutilide). clofilium 186-195 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 15266014-12 2004 In summary, the pore helix residues are important components of the HERG drug binding site, and may be particularly important for drugs with polar substituents, such as a halogen (e.g., clofilium) or a methanesulfonamide (e.g., ibutilide). methanesulfonamide 202-220 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 15266014-12 2004 In summary, the pore helix residues are important components of the HERG drug binding site, and may be particularly important for drugs with polar substituents, such as a halogen (e.g., clofilium) or a methanesulfonamide (e.g., ibutilide). ibutilide 228-237 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 15322737-4 2004 Therefore, we investigated the effect of trazodone on HERG potassium channels expressed in human embryonic kidney (HEK) cells and in Xenopus oocytes. Trazodone 41-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 15322737-5 2004 Trazodone inhibited HERG currents in a dose-dependent manner with an IC50 of 2.9 microM in HEK cells and 13.2 microM in Xenopus oocytes. Trazodone 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 15322737-7 2004 In HERG channel mutants Y652A and F656A lacking aromatic residues in the S6 domain, the affinity of trazodone was reduced profoundly. Trazodone 100-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 15322737-8 2004 Trazodone accelerated inactivation of HERG currents without markedly affecting activation. Trazodone 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 15322737-11 2004 Trazodone block of HERG channels was state dependent. Trazodone 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 15322737-13 2004 In summary, the atypical antidepressant trazodone blocks cardiac HERG channels at concentrations that are probably relevant in vivo, particularly in overdosage. Trazodone 40-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 15189761-0 2004 Comparison of kinetic properties of quinidine and dofetilide block of HERG channels. Quinidine 36-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 14586519-2 2004 Inward HERG currents were recorded on hyperpolarization in 140 mM external Cs(+) and Rb(+), as well as K(+). Cesium 75-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 7-11 14586519-2 2004 Inward HERG currents were recorded on hyperpolarization in 140 mM external Cs(+) and Rb(+), as well as K(+). Rubidium 85-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 7-11 15189761-0 2004 Comparison of kinetic properties of quinidine and dofetilide block of HERG channels. dofetilide 50-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 15189761-2 2004 We examined the kinetic properties of quinidine block of HERG channels expressed in Xenopus oocytes in comparison with those of the block by a class III antiarrhythmic dofetilide. Quinidine 38-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 15189761-8 2004 In contrast, dofetilide blocked HERG currents in a voltage- and time-independent manner under the steady state because of very slow unblocking at negative potentials, which also caused frequency-independent block. dofetilide 13-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 14975928-12 2004 All HERG channels had similar sensitivity to block by cisapride. Cisapride 54-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 15148258-0 2004 Inhibition of cardiac HERG currents by the DNA topoisomerase II inhibitor amsacrine: mode of action. Amsacrine 74-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 15148258-3 2004 Since blockade of cardiac human ether-a-go-go-related gene (HERG) potassium currents is an important cause of acquired LQTS, we investigated the acute effects of amsacrine on cloned HERG channels to determine the electrophysiological basis for its proarrhythmic potential. Potassium 66-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 15148258-4 2004 2 HERG channels were heterologously expressed in human HEK 293 cells and Xenopus laevis oocytes, and the respective potassium currents were recorded using patch-clamp and two-microelectrode voltage-clamp electrophysiology. Potassium 116-125 potassium voltage-gated channel subfamily H member 2 Homo sapiens 2-6 15148258-5 2004 3 Amsacrine blocked HERG currents in HEK 293 cells and Xenopus oocytes in a concentration-dependent manner, with IC50 values of 209.4 nm and 2.0 microm, respectively. Amsacrine 2-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 15148258-8 2004 HERG current block by amsacrine was not frequency dependent. Amsacrine 22-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 15148258-9 2004 5 The S6 domain mutations Y652A and F656A attenuated (Y652A) or abolished (F656A, Y652A/F656A) HERG current blockade, indicating that amsacrine binding requires a common drug receptor within the pore-S6 region. Amsacrine 134-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 95-99 15148258-10 2004 6 In conclusion, these data demonstrate that the anticancer drug amsacrine is an antagonist of cloned HERG potassium channels, providing a molecular mechanism for the previously reported QTc interval prolongation during clinical administration of amsacrine. Amsacrine 65-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 15148258-10 2004 6 In conclusion, these data demonstrate that the anticancer drug amsacrine is an antagonist of cloned HERG potassium channels, providing a molecular mechanism for the previously reported QTc interval prolongation during clinical administration of amsacrine. Amsacrine 247-256 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 14744814-0 2004 Potential mechanisms for the enhancement of HERG K+ channel function by phospholipid metabolites. Phospholipids 72-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15272206-0 2004 Validation of a [3H]astemizole binding assay in HEK293 cells expressing HERG K+ channels. Tritium 17-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 15071359-0 2004 Block of HERG human K(+) channel and IKr of guinea pig cardiomyocytes by chlorpromazine. Chlorpromazine 73-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 15071359-2 2004 We studied the effects of chlorpromazine on the human ether-a-go-go-related gene (HERG) channel expressed in Xenopus oocytes and on delayed rectifier K current of guinea pig ventricular myocytes. Chlorpromazine 26-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 15071359-3 2004 Application of chlorpromazine showed a dose-dependent decrease in the amplitudes of steady-state currents and tail currents of HERG. Chlorpromazine 15-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 15071359-4 2004 The decrease became more pronounced at increasingly positive potential, suggesting that the blockade of HERG by chlorpromazine is voltage dependent. Chlorpromazine 112-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 15071359-5 2004 IC50 for chlorpromazine block of HERG current was progressively decreased according to depolarization: IC50 values at -30, 0, and +30 mV were 10.5, 8.8, and 4.9 microM, respectively. Chlorpromazine 9-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 15071359-9 2004 Our findings suggest that the arrhythmogenic side effect of chlorpromazine is caused by blockade of HERG and rapid component of delayed rectifier K current rather than by blockade of the slow component. Chlorpromazine 60-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 15272206-0 2004 Validation of a [3H]astemizole binding assay in HEK293 cells expressing HERG K+ channels. Astemizole 20-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 15272206-2 2004 Pharmacological characterization of the [(3)H]astemizole binding assay for HERG K(+) channels was performed using HERG-expressing HEK293 cells. Astemizole 46-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 75-79 15272206-2 2004 Pharmacological characterization of the [(3)H]astemizole binding assay for HERG K(+) channels was performed using HERG-expressing HEK293 cells. Astemizole 46-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 15272206-5 2004 Binding affinities for 32 reference compounds (including dofetilide, cisapride, and terfenadine) with diverse structures demonstrated a similar potency rank order for HERG inhibition to that reported in the literature. dofetilide 57-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 167-171 15272206-5 2004 Binding affinities for 32 reference compounds (including dofetilide, cisapride, and terfenadine) with diverse structures demonstrated a similar potency rank order for HERG inhibition to that reported in the literature. Cisapride 69-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 167-171 15272206-5 2004 Binding affinities for 32 reference compounds (including dofetilide, cisapride, and terfenadine) with diverse structures demonstrated a similar potency rank order for HERG inhibition to that reported in the literature. Terfenadine 84-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 167-171 15272206-6 2004 Moreover, the [(3)H]astemizole binding data demonstrated a rank order of affinity that was highly correlated to that of inhibitory potency in the electrophysiological studies for HERG in HEK293 (r(SP) = 0.91, P<0.05). Tritium 14-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 15272206-6 2004 Moreover, the [(3)H]astemizole binding data demonstrated a rank order of affinity that was highly correlated to that of inhibitory potency in the electrophysiological studies for HERG in HEK293 (r(SP) = 0.91, P<0.05). Astemizole 20-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 15272206-7 2004 In conclusion, the [(3)H]astemizole binding assay is rapid and capable of detecting HERG inhibitors. Astemizole 25-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 15120636-0 2004 Potent inhibition of human cardiac potassium (HERG) channels by the anti-estrogen agent clomiphene-without QT interval prolongation. Clomiphene 88-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 15120636-3 2004 We describe here a potent inhibitory effect (IC(50) = 0.18 microM) of clomiphene on HERG ionic current (I(HERG)) recorded from a mammalian cell line expressing HERG channels. Clomiphene 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 15120636-3 2004 We describe here a potent inhibitory effect (IC(50) = 0.18 microM) of clomiphene on HERG ionic current (I(HERG)) recorded from a mammalian cell line expressing HERG channels. Clomiphene 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 15120636-3 2004 We describe here a potent inhibitory effect (IC(50) = 0.18 microM) of clomiphene on HERG ionic current (I(HERG)) recorded from a mammalian cell line expressing HERG channels. Clomiphene 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 15120636-4 2004 Inhibition of I(HERG) by clomiphene showed voltage-dependence and developed quickly following membrane depolarisation, indicating contingency of block on HERG channel gating. Clomiphene 25-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 15120636-4 2004 Inhibition of I(HERG) by clomiphene showed voltage-dependence and developed quickly following membrane depolarisation, indicating contingency of block on HERG channel gating. Clomiphene 25-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 154-158 15120636-5 2004 At 100 nM, clomiphene and the related anti-estrogen tamoxifen produced similar levels of I(HERG) blockade (p > 0.05). Clomiphene 11-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 15120636-5 2004 At 100 nM, clomiphene and the related anti-estrogen tamoxifen produced similar levels of I(HERG) blockade (p > 0.05). Tamoxifen 52-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 15120636-7 2004 The disparity between clomiphene"s potent I(HERG) inhibition and its lack of effect on the QT interval underscores the notion that I(HERG) pharmacology may best be used alongside other screening methods when investigating the QT-prolonging tendency and related cardiotoxicity of non-cardiac drugs. Clomiphene 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15120636-7 2004 The disparity between clomiphene"s potent I(HERG) inhibition and its lack of effect on the QT interval underscores the notion that I(HERG) pharmacology may best be used alongside other screening methods when investigating the QT-prolonging tendency and related cardiotoxicity of non-cardiac drugs. Clomiphene 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 15125690-2 2004 The ether-a-go-go-related gene, HERG, is a primary target for blockade by many drugs including dofetilide, quinidine and azimilide. dofetilide 95-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 15125690-2 2004 The ether-a-go-go-related gene, HERG, is a primary target for blockade by many drugs including dofetilide, quinidine and azimilide. Quinidine 107-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 15125690-2 2004 The ether-a-go-go-related gene, HERG, is a primary target for blockade by many drugs including dofetilide, quinidine and azimilide. azimilide 121-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 15125690-10 2004 Acidification weakened the inhibitory effects of quinidine and azimilide on HERG channels. Quinidine 49-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 15125690-10 2004 Acidification weakened the inhibitory effects of quinidine and azimilide on HERG channels. azimilide 63-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 15125690-12 2004 Acidification markedly potentiated dofetilide blockade of the HERG channels but weakened the inhibitory effects of quinidine and azimilide. dofetilide 35-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 15168062-0 2004 Block of HERG current expressed in HEK293 cells by the Na+-channel blocker cibenzoline. cifenline 75-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 15168062-1 2004 A Na(+)-channel blocker, cibenzoline, blocks the delayed rectifier potassium current ( I(k)), but its detailed action on the rapidly activating component ( I(kr)) of I(k) encoded by the human ether-a-go-go-related gene ( HERG) has not been clarified. cifenline 25-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 221-225 15168062-2 2004 We examined the effects of cibenzoline on stably expressed HERG current in HEK293 cells recorded by the patch-clamp technique of whole-cell configuration. cifenline 27-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 15168062-3 2004 Cibenzoline blocked HERG current expressed in HEK293 cells with IC(50) = 3.7 +/- 0.963 micro M and Hill coefficient = 0.74 +/- 0.12. cifenline 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 15168062-9 2004 Cibenzoline blocks the I(kr)-like current reconstituted by HERG clone transfection with an IC(50) value comparable to therapeutic concentrations. cifenline 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 15168062-10 2004 Cibenzoline has a preferential affinity, at least, to the open state of the HERG channel with a rapid access to the binding site. cifenline 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 15135665-0 2004 Lidoflazine is a high affinity blocker of the HERG K(+)channel. Lidoflazine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 15135665-3 2004 Lidoflazine inhibited potently HERG current (I(HERG)) recorded from HEK 293 cells stably expressing wild-type HERG (IC(50) of approximately 16 nM). Lidoflazine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 15135665-3 2004 Lidoflazine inhibited potently HERG current (I(HERG)) recorded from HEK 293 cells stably expressing wild-type HERG (IC(50) of approximately 16 nM). Lidoflazine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 15135665-3 2004 Lidoflazine inhibited potently HERG current (I(HERG)) recorded from HEK 293 cells stably expressing wild-type HERG (IC(50) of approximately 16 nM). Lidoflazine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 15135665-6 2004 The effect of command voltage on the drug"s action suggested that lidoflazine preferentially inhibits activated/open HERG channels. Lidoflazine 66-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 15135665-8 2004 We conclude: first, that lidoflazine produces high affinity blockade of the alpha subunit of the HERG channel by binding to aromatic amino acid residues within the channel pore and, second, that this is likely to represent the molecular mechanism of QT interval prolongation by this drug. Lidoflazine 25-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 15135665-8 2004 We conclude: first, that lidoflazine produces high affinity blockade of the alpha subunit of the HERG channel by binding to aromatic amino acid residues within the channel pore and, second, that this is likely to represent the molecular mechanism of QT interval prolongation by this drug. Amino Acids, Aromatic 124-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 15102940-4 2004 Clofilium inhibited hEAG1 and hERG1 with the same potency, whereas hEAG2 was about 150-fold less sensitive to this antiarrhythmic agent. clofilium 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-35 15098086-7 2004 To assess the action of the effects of prazosin, doxazosin, and terazosin on HERG currents, we investigated their acute electrophysiological effects on cloned HERG potassium channels heterologously expressed in Xenopus oocytes and HEK 293 cells.Prazosin, doxazosin, and terazosin blocked HERG currents in Xenopus oocytes with IC(50) values of 10.1, 18.2, and 113.2 microM respectively, whereas the IC(50) values for HERG channel inhibition in human HEK 293 cells were 1.57 microM, 585.1 nM, and 17.7 microM. Terazosin 64-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 14711935-4 2004 Using patch-clamp electrophysiology, we found that tolterodine was a potent antagonist of the human ether-a-go-go-related gene (HERG) K(+) channel, displaying an IC(50) value of 17 nM. Tolterodine Tartrate 51-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 14711935-6 2004 Tolterodine block of HERG displayed a positive voltage dependence, suggesting an interaction with an activated state. Tolterodine Tartrate 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 14711935-11 2004 Tolterodine seems to be an unusual drug in that it blocks HERG with high affinity, but produces little QT prolongation clinically. Tolterodine Tartrate 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 15098086-12 2004 The S6 mutations Y652A and F656A partially attenuated (Y652A) or abolished (F656A) HERG current blockade, indicating that prazosin binds to a common drug receptor within the pore-S6 region. Prazosin 122-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 15098086-13 2004 In conclusion, this study demonstrates that HERG potassium channels are blocked by prazosin, doxazosin, and terazosin. Prazosin 83-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15098086-13 2004 In conclusion, this study demonstrates that HERG potassium channels are blocked by prazosin, doxazosin, and terazosin. Doxazosin 93-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 15098086-13 2004 In conclusion, this study demonstrates that HERG potassium channels are blocked by prazosin, doxazosin, and terazosin. Terazosin 108-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 14744814-3 2004 Here we studied effects of several lysophospholipids with different lengths of hydrocarbon chains and charged headgroups on HERG K(+) currents (I(HERG)) expressed in HEK293 cells and the potential mechanisms using whole-cell patch-clamp techniques. Lysophospholipids 35-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 14744814-3 2004 Here we studied effects of several lysophospholipids with different lengths of hydrocarbon chains and charged headgroups on HERG K(+) currents (I(HERG)) expressed in HEK293 cells and the potential mechanisms using whole-cell patch-clamp techniques. Lysophospholipids 35-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 146-150 14744814-5 2004 Only the lipids with 16 hydrocarbons such as 1-palmitoyl-lysophosphatidylcholine (LPC-16) and 1-palmitoyl-lysophosphatidylglycerol (LPG-16) were found to produce significant enhancement of I(HERG) and negative shifts of HERG activation, although the voltage dependence of the effects was different between LPC-16 and LPG-16 which have differently charged headgroups. We 201 45-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 191-195 14744814-5 2004 Only the lipids with 16 hydrocarbons such as 1-palmitoyl-lysophosphatidylcholine (LPC-16) and 1-palmitoyl-lysophosphatidylglycerol (LPG-16) were found to produce significant enhancement of I(HERG) and negative shifts of HERG activation, although the voltage dependence of the effects was different between LPC-16 and LPG-16 which have differently charged headgroups. We 201 45-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 220-224 14744814-5 2004 Only the lipids with 16 hydrocarbons such as 1-palmitoyl-lysophosphatidylcholine (LPC-16) and 1-palmitoyl-lysophosphatidylglycerol (LPG-16) were found to produce significant enhancement of I(HERG) and negative shifts of HERG activation, although the voltage dependence of the effects was different between LPC-16 and LPG-16 which have differently charged headgroups. 1-palmitoyl-lysophosphatidylglycerol 94-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 191-195 14744814-5 2004 Only the lipids with 16 hydrocarbons such as 1-palmitoyl-lysophosphatidylcholine (LPC-16) and 1-palmitoyl-lysophosphatidylglycerol (LPG-16) were found to produce significant enhancement of I(HERG) and negative shifts of HERG activation, although the voltage dependence of the effects was different between LPC-16 and LPG-16 which have differently charged headgroups. 1-palmitoyl-lysophosphatidylglycerol 94-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 220-224 14744814-6 2004 The lipid with 18 hydrocarbons modestly increased I(HERG). Hydrocarbons 18-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 14744814-14 2004 We conclude that enhancement of HERG function by lysophospholipids is specific to the lipids with 16-hydrocarbon chain structure and the pattern of voltage dependence is determined by the polar headgroups. Lysophospholipids 49-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 15176423-2 2004 Inhibition of HERG potassium currents by class III antiarrhythmic drugs causes lengthening of the cardiac action potential, which produces a beneficial antiarrhythmic effect. Potassium 19-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 14709917-5 2004 Terfenadine 1 microM completely inhibited the human ether a go-go-related gene (HERG) channel current expressed in HEK293 cells in the same experimental solution as in microelectrode experiments. Terfenadine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 14665560-8 2004 RESULTS: The amide local anaesthetics bupivacaine, levobupivacaine and ropivacaine inhibited HERG channels at toxicologically relevant concentrations, with IC(50) values of 20 (SEM 2) micro M (n=29), 10 (1) micro M (n=40) and 20 (2) micro M (n=49), respectively. Amides 13-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 14665560-8 2004 RESULTS: The amide local anaesthetics bupivacaine, levobupivacaine and ropivacaine inhibited HERG channels at toxicologically relevant concentrations, with IC(50) values of 20 (SEM 2) micro M (n=29), 10 (1) micro M (n=40) and 20 (2) micro M (n=49), respectively. Bupivacaine 38-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 14665560-8 2004 RESULTS: The amide local anaesthetics bupivacaine, levobupivacaine and ropivacaine inhibited HERG channels at toxicologically relevant concentrations, with IC(50) values of 20 (SEM 2) micro M (n=29), 10 (1) micro M (n=40) and 20 (2) micro M (n=49), respectively. Levobupivacaine 51-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 14665560-8 2004 RESULTS: The amide local anaesthetics bupivacaine, levobupivacaine and ropivacaine inhibited HERG channels at toxicologically relevant concentrations, with IC(50) values of 20 (SEM 2) micro M (n=29), 10 (1) micro M (n=40) and 20 (2) micro M (n=49), respectively. Ropivacaine 71-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 14665560-13 2004 CONCLUSIONS: Amide local anaesthetics target HERG and HERG/MiRP1 channels with identical potency. Amides 13-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 14665560-13 2004 CONCLUSIONS: Amide local anaesthetics target HERG and HERG/MiRP1 channels with identical potency. Amides 13-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 15107589-0 2004 HERG K channel conductance promotes H2O2-induced apoptosis in HEK293 cells: cellular mechanisms. Hydrogen Peroxide 36-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 15107589-9 2004 HERG expression facilitates DNA fragmentation induced by H(2)O(2) at a concentration-dependent fashion, starting at 200 microM and reaching maximum at 1 mM. Hydrogen Peroxide 57-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 15107589-11 2004 Inhibition of p38 by SB-203580 alleviated DNA-F and PD-98059, which inhibited activation of ERKs, nearly abolished DNA-F. Immunoblotting analysis demonstrated that p38, SAPKs and ERKs MAP kinases were all substantially activated (>10-fold higher) in HERG-expressing cells vs. non-transfected cells. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 52-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 253-257 14744814-14 2004 We conclude that enhancement of HERG function by lysophospholipids is specific to the lipids with 16-hydrocarbon chain structure and the pattern of voltage dependence is determined by the polar headgroups. Hydrocarbons 101-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 14646167-4 2003 In conclusion, these evaluation methods demonstrated that ER-118585 could prolong the QT interval via APD prolongation, attributable to the inhibition of the HERG potassium current. Potassium 163-172 potassium voltage-gated channel subfamily H member 2 Homo sapiens 158-162 15371638-3 2004 Within those segments thought to contribute to the channel pore, HERG possesses several serine residues that are not present in EAG channels. Serine 88-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 15371638-6 2004 As with the other serines, S641 is also involved in maintaining ion selectivity of the HERG channel and alters sensitivity to block by E4031. Serine 18-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 15371638-6 2004 As with the other serines, S641 is also involved in maintaining ion selectivity of the HERG channel and alters sensitivity to block by E4031. 3-Methoxydiphenylamine 27-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 15371638-6 2004 As with the other serines, S641 is also involved in maintaining ion selectivity of the HERG channel and alters sensitivity to block by E4031. E 4031 135-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 14734057-2 2004 In this study, we examined the effects of mesoridazine on human ether-a-go-go-related gene (HERG) K+ currents. Mesoridazine 42-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 14734057-4 2004 Mesoridazine blocked HERG currents in a concentration-dependent manner (IC50 550 nM at 0 mV); block increased significantly over the voltage range where HERG activates and saturated at voltages eliciting maximal HERG channel activation. Mesoridazine 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 14734057-4 2004 Mesoridazine blocked HERG currents in a concentration-dependent manner (IC50 550 nM at 0 mV); block increased significantly over the voltage range where HERG activates and saturated at voltages eliciting maximal HERG channel activation. Mesoridazine 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 153-157 14734057-4 2004 Mesoridazine blocked HERG currents in a concentration-dependent manner (IC50 550 nM at 0 mV); block increased significantly over the voltage range where HERG activates and saturated at voltages eliciting maximal HERG channel activation. Mesoridazine 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 153-157 14734057-5 2004 Tonic block of HERG current by mesoridazine (1.8 microM) was minimal (< 2-4%). Mesoridazine 31-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 14734057-6 2004 The rate of the onset of HERG channel block was rapid and dose dependent (tau = 54 +/- 7 ms at 0 mV and 1.8 microM mesoridazine), but not significantly affected by test potentials ranging from -30 to +30 mV. Mesoridazine 115-127 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 14734057-10 2004 These findings demonstrate that mesoridazine is a potent and rapid open-channel blocker of HERG channels. Mesoridazine 32-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 14525949-6 2003 Functionally, KCR1 reduces the sensitivity of HERG to classic proarrhythmic HERG blockers (sotalol, quinidine, dofetilide) in both cardiac and noncardiac cell lines. Sotalol 91-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 14525949-6 2003 Functionally, KCR1 reduces the sensitivity of HERG to classic proarrhythmic HERG blockers (sotalol, quinidine, dofetilide) in both cardiac and noncardiac cell lines. Sotalol 91-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 14525949-6 2003 Functionally, KCR1 reduces the sensitivity of HERG to classic proarrhythmic HERG blockers (sotalol, quinidine, dofetilide) in both cardiac and noncardiac cell lines. Quinidine 100-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 14525949-6 2003 Functionally, KCR1 reduces the sensitivity of HERG to classic proarrhythmic HERG blockers (sotalol, quinidine, dofetilide) in both cardiac and noncardiac cell lines. dofetilide 111-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 14525949-6 2003 Functionally, KCR1 reduces the sensitivity of HERG to classic proarrhythmic HERG blockers (sotalol, quinidine, dofetilide) in both cardiac and noncardiac cell lines. dofetilide 111-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 14674677-0 2003 Characterization of the inhibitory effects of erythromycin and clarithromycin on the HERG potassium channel. Erythromycin 46-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 14642687-0 2003 A new oral therapy for long QT syndrome: long-term oral potassium improves repolarization in patients with HERG mutations. Potassium 56-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-111 14642687-1 2003 OBJECTIVES: We sought to determine whether oral potassium supplementation safely increases serum K(+) and results in sustained improvement of repolarization parameters in long QT syndrome type 2 (LQT2) subjects. Potassium 48-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 196-200 14642687-4 2003 We tested the hypothesis that long-term oral potassium supplementation results in a mild, sustainable increase in serum K(+) that improves repolarization abnormalities in subjects with LQT2. Potassium 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 14642687-13 2003 CONCLUSIONS: Long-term oral potassium administration increases serum K(+) in patients with LQT2. Potassium 28-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 14674677-0 2003 Characterization of the inhibitory effects of erythromycin and clarithromycin on the HERG potassium channel. Clarithromycin 63-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 14674677-2 2003 The goal of this study was to test the hypothesis that these macrolide antibiotics significantly block the delayed rectifier current (IKr) encoded by HERG (the human ether-a-go-go-related gene) at drug concentrations, temperature and ionic conditions mimicking those occurring in human subjects. Macrolides 61-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 150-154 14674677-3 2003 Potassium currents in HEK 293 cells stably transfected with HERG were recorded using a whole cell voltage clamp method. Potassium 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 14674677-4 2003 Exposure of cells to erythromycin reduced the HERG encoded potassium current in a concentration dependent manner with an IC50 of 38.9 +/- 1.2 microM and Hill Slope factor of 0.4 +/- 0.1. Erythromycin 21-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 14674677-4 2003 Exposure of cells to erythromycin reduced the HERG encoded potassium current in a concentration dependent manner with an IC50 of 38.9 +/- 1.2 microM and Hill Slope factor of 0.4 +/- 0.1. Potassium 59-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 14674677-7 2003 The results of this study document that both erythromycin and clarithromycin significantly inhibit the HERG potassium current at clinically relevant concentrations. Erythromycin 45-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 14674677-7 2003 The results of this study document that both erythromycin and clarithromycin significantly inhibit the HERG potassium current at clinically relevant concentrations. Clarithromycin 62-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 14674677-7 2003 The results of this study document that both erythromycin and clarithromycin significantly inhibit the HERG potassium current at clinically relevant concentrations. Potassium 108-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 14576514-6 2003 Both drugs blocked activated HERG channels in a biexponential decay fashion, with faster time constants for KCB-328 (3 microM) than for dofetilide (0.3 microM). dofetilide 136-146 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 14557918-0 2003 Drug binding to aromatic residues in the HERG channel pore cavity as possible explanation for acquired Long QT syndrome by antiparkinsonian drug budipine. budipine 145-153 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 14557918-6 2003 In Xenopus oocytes, HERG potassium channels were blocked by budipine with an IC(50) of 10.2 microM. budipine 60-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 14557918-8 2003 Budipine blocked HERG channels in the open and inactivated state, but not in the closed states. budipine 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 14557918-10 2003 Steady-state inactivation of HERG was also influenced by budipine. budipine 57-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 12902341-5 2003 In this study we show that a synthetic 42-residue peptide corresponding to this linker region of the HERG K+ channel does not have defined structural elements in aqueous solution; however, it displays two well defined helical regions when in the presence of SDS micelles. Sodium Dodecyl Sulfate 258-261 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 12885765-8 2003 This study reveals a novel role of the COOH terminus in the normal biogenesis of HERG channels and suggests defective trafficking as a common mechanism for abnormal channel function resulting from mutations of critical COOH-terminal residues, including the LQTS mutant HERGN861I. Carbonic Acid 39-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 15326914-3 2003 Tertiary amines which form ammonium ions shielded by two structural fragments of the drug molecule were found to be potent HERG/I(Kr) blockers with IC50 < 1 microM (16 of 19 compounds, 84%). Amines 9-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 123-127 15326914-3 2003 Tertiary amines which form ammonium ions shielded by two structural fragments of the drug molecule were found to be potent HERG/I(Kr) blockers with IC50 < 1 microM (16 of 19 compounds, 84%). Ammonium Compounds 27-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 123-127 15326914-5 2003 Similarly, 27 of 32 weak HERG blockers ( IC50 > 10 microM) were found to be primary or secondary amines, or neutral or very weakly basic compounds. Amines 100-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 15326914-7 2003 Conformational analysis and modeling of the interaction of the charged fragment of the drugs with acetone, a system that mimics a ketone fragment of HERG/I(Kr) channel, supports preference of the conformation with the shielded charged center for potent HERG/I(Kr) blockers. Acetone 98-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 15326914-7 2003 Conformational analysis and modeling of the interaction of the charged fragment of the drugs with acetone, a system that mimics a ketone fragment of HERG/I(Kr) channel, supports preference of the conformation with the shielded charged center for potent HERG/I(Kr) blockers. Acetone 98-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 253-257 15326914-7 2003 Conformational analysis and modeling of the interaction of the charged fragment of the drugs with acetone, a system that mimics a ketone fragment of HERG/I(Kr) channel, supports preference of the conformation with the shielded charged center for potent HERG/I(Kr) blockers. Ketones 130-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 15326914-7 2003 Conformational analysis and modeling of the interaction of the charged fragment of the drugs with acetone, a system that mimics a ketone fragment of HERG/I(Kr) channel, supports preference of the conformation with the shielded charged center for potent HERG/I(Kr) blockers. Ketones 130-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 253-257 15326914-9 2003 We suggest that the introduction of a hydroxy group at position 3 relative to a tertiary ammonium charged center, or the introduction of hydroxy, alkoxy or amino groups at position 2 relative to the nitrogen center of an aromatic system, should provide easy access of a water molecule to the proton, thereby facilitating deprotonation and thus leading to a moderate or weak HERG/I(Kr) blockade and a reduced risk of TdP. Water 270-275 potassium voltage-gated channel subfamily H member 2 Homo sapiens 374-378 14527710-6 2003 methadone causes QTc prolongation in humans; (2) whether methadone and/or chlorobutanol block cardiac HERG potassium currents (IHERG) in vitro. Methadone 57-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 14527710-6 2003 methadone causes QTc prolongation in humans; (2) whether methadone and/or chlorobutanol block cardiac HERG potassium currents (IHERG) in vitro. Chlorobutanol 74-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 14527710-6 2003 methadone causes QTc prolongation in humans; (2) whether methadone and/or chlorobutanol block cardiac HERG potassium currents (IHERG) in vitro. Potassium 107-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 12837749-0 2003 Thapsigargin selectively rescues the trafficking defective LQT2 channels G601S and F805C. Thapsigargin 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 12837749-12 2003 This study 1) supports the hypothesis that the LQT2 trafficking defective phenotype can be reversed without blocking the channel; 2) demonstrates pharmacological rescue of a C terminus LQT2 mutation; and 3) shows that thapsigargin can correct trafficking defective phenotypes in more than one channel type and disease (i.e. LQT2 and cystic fibrosis). Thapsigargin 218-230 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 12837749-12 2003 This study 1) supports the hypothesis that the LQT2 trafficking defective phenotype can be reversed without blocking the channel; 2) demonstrates pharmacological rescue of a C terminus LQT2 mutation; and 3) shows that thapsigargin can correct trafficking defective phenotypes in more than one channel type and disease (i.e. LQT2 and cystic fibrosis). Thapsigargin 218-230 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 12837749-12 2003 This study 1) supports the hypothesis that the LQT2 trafficking defective phenotype can be reversed without blocking the channel; 2) demonstrates pharmacological rescue of a C terminus LQT2 mutation; and 3) shows that thapsigargin can correct trafficking defective phenotypes in more than one channel type and disease (i.e. LQT2 and cystic fibrosis). Thapsigargin 218-230 potassium voltage-gated channel subfamily H member 2 Homo sapiens 185-189 14512100-0 2003 Prulifloxacin: in vitro (HERG current) and in vivo (conscious dog) assessment of cardiac risk. prulifloxacin 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 14512100-1 2003 Prulifloxacin, a new thiazeto-quinoline derivative with antibiotic properties, was evaluated for cardiac risk both in vitro on the ether-a-go-go-related gene (HERG) K+ channel, and in vivo in the conscious dog monitored by telemetry. prulifloxacin 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 159-163 14512100-5 2003 In contrast, moxifloxacin blocked HERG current amplitude with an IC50 value of 74.7 microM. Moxifloxacin 13-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 12960687-0 2003 Effects of Na+ channel blocker, pilsicainide, on HERG current expressed in HEK-293 cells. pilsicainide 32-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 14499861-1 2003 OBJECTIVE: To determine whether the amino acid 897 threonine (T) to lysine (K) polymorphism of the KCNH2 (HERG) potassium channel influences channel performance or patient phenotype. Threonine 51-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-104 14499861-1 2003 OBJECTIVE: To determine whether the amino acid 897 threonine (T) to lysine (K) polymorphism of the KCNH2 (HERG) potassium channel influences channel performance or patient phenotype. Threonine 51-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 14499861-1 2003 OBJECTIVE: To determine whether the amino acid 897 threonine (T) to lysine (K) polymorphism of the KCNH2 (HERG) potassium channel influences channel performance or patient phenotype. Lysine 68-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-104 14499861-1 2003 OBJECTIVE: To determine whether the amino acid 897 threonine (T) to lysine (K) polymorphism of the KCNH2 (HERG) potassium channel influences channel performance or patient phenotype. Lysine 68-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 12960687-2 2003 We studied effects of pilsicainide on the K+ channel current of the human ether-a-go-go-related gene (HERG) in heterologous expression system. pilsicainide 22-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 12960687-4 2003 RESULTS: Pilsicainide suppressed peak currents of HERG channel during depolarizing pulses and tail currents upon repolarization. pilsicainide 9-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 12960687-5 2003 Pilsicainide blocked HERG current with IC50 = 20.4 microM and Hill coefficient = 0.98. pilsicainide 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 12960687-11 2003 CONCLUSIONS: Pilsicainide blocks HERG current with a preferential affinity, at least, to the open state of the channels with a fast access to binding sites. pilsicainide 13-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 12860380-0 2003 Preferential closed channel blockade of HERG potassium currents by chemically synthesised BeKm-1 scorpion toxin. Potassium 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 12860380-5 2003 Blockade by BeKm-1 and recombinant ergtoxin, another scorpion toxin known to block HERG, differed in their recovery from HERG current inactivation elicited by strong depolarisation and in their ability to block HERG when the channels were already activated. (6aR,9R)-N-[(1S,2S,4R,7S)-7-benzyl-2-hydroxy-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-5,8-dioxo-4,7-di(propan-2-yl)-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-7-(2-methylpropyl)-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide 35-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 12860380-5 2003 Blockade by BeKm-1 and recombinant ergtoxin, another scorpion toxin known to block HERG, differed in their recovery from HERG current inactivation elicited by strong depolarisation and in their ability to block HERG when the channels were already activated. (6aR,9R)-N-[(1S,2S,4R,7S)-7-benzyl-2-hydroxy-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-5,8-dioxo-4,7-di(propan-2-yl)-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-7-(2-methylpropyl)-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide 35-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 12860380-5 2003 Blockade by BeKm-1 and recombinant ergtoxin, another scorpion toxin known to block HERG, differed in their recovery from HERG current inactivation elicited by strong depolarisation and in their ability to block HERG when the channels were already activated. (6aR,9R)-N-[(1S,2S,4R,7S)-7-benzyl-2-hydroxy-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-5,8-dioxo-4,7-di(propan-2-yl)-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-7-(2-methylpropyl)-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide 35-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 12839860-0 2003 Drug binding to HERG channels: evidence for a "non-aromatic" binding site for fluvoxamine. Fluvoxamine 78-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 12839862-0 2003 Blockade of HERG potassium currents by fluvoxamine: incomplete attenuation by S6 mutations at F656 or Y652. Potassium 17-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 12839862-0 2003 Blockade of HERG potassium currents by fluvoxamine: incomplete attenuation by S6 mutations at F656 or Y652. Fluvoxamine 39-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 12839862-3 2003 The objectives of this study were (i) to identify and characterise any inhibitory action on HERG of the selective-serotonin re-uptake inhibitor fluvoxamine, (ii) to then determine whether fluvoxamine shared the consensus molecular determinants of HERG blockade of those drugs so far tested. Fluvoxamine 144-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 12839862-6 2003 I(HERG) tails, following repolarisation from +20 to -40 mV, were blocked by fluvoxamine with an IC(50) of 3.8 micro M. 3. Fluvoxamine 76-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 2-6 12839862-7 2003 Blockade of wild-type HERG was of extremely rapid onset (within 10 ms) and showed voltage dependence, with fluvoxamine also inducing a leftward shift in voltage-dependent activation of I(HERG). Fluvoxamine 107-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 12839862-7 2003 Blockade of wild-type HERG was of extremely rapid onset (within 10 ms) and showed voltage dependence, with fluvoxamine also inducing a leftward shift in voltage-dependent activation of I(HERG). Fluvoxamine 107-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 187-191 12839862-11 2003 The S6 mutations, Y652A and F656A, and the pore helix mutant S631A only partially attenuated blockade by fluvoxamine at concentrations causing profound blockade of wild-type HERG. Fluvoxamine 105-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 12804575-0 2003 Characterisation of recombinant HERG K+ channel blockade by the Class Ia antiarrhythmic drug procainamide. Procainamide 93-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 14510655-8 2003 CONCLUSIONS: Phenylephrine-induced bradycardia decreased TDR in symptomatic LQT1 but increased TDR in symptomatic LQT2. Phenylephrine 13-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 12829172-6 2003 RESULTS: Application of the phorbol ester PMA, an unspecific protein kinase activator, shifted the voltage dependence of HERG activation towards more positive potentials. Phorbol Esters 28-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 12827215-0 2003 Inhibition of cloned HERG potassium channels by the antiestrogen tamoxifen. Tamoxifen 65-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 12827215-5 2003 Tamoxifen blocked HERG potassium channels with an IC(50) value of 45.3 microM. Tamoxifen 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 12827215-9 2003 This study demonstrates that HERG potassium channels are blocked by the antiestrogenic drug tamoxifen. Tamoxifen 92-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 12775586-5 2003 The specific Hsp90 inhibitor geldanamycin prevents maturation and increases proteasomal degradation of hERG WT, while reducing hERG currents in heterologous expression systems. geldanamycin 29-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 12759137-5 2003 We favour the latter hypothesis, since some fluoroquinolones have recently been shown to block the pore-forming subunit of the cardiac rapid delayed rectifier K(+) current I(Kr) (which is encoded by HERG (human ether-a-go-go-related gene)). Fluoroquinolones 44-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 199-203 12740430-0 2003 Inhibition of HERG K+ current and prolongation of the guinea-pig ventricular action potential by 4-aminopyridine. 4-Aminopyridine 97-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 12740430-2 2003 We report here inhibition by 4-AP of HERG (the human ether-a-go-go-related gene) K+ channels expressed in a mammalian cell line, at concentrations relevant to those used to study ITO,1. 4-Aminopyridine 29-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 12736144-9 2003 Low concentrations of the KCNH2 blockers E-4031 (10(-8) M) and MK-499 (3 x 10(-8) M) increased phasic contractile amplitude and the number of spikes per slow wave. E 4031 41-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-31 12736144-9 2003 Low concentrations of the KCNH2 blockers E-4031 (10(-8) M) and MK-499 (3 x 10(-8) M) increased phasic contractile amplitude and the number of spikes per slow wave. L 706000 63-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-31 12788816-0 2003 The antipsychotic drug chlorpromazine inhibits HERG potassium channels. Chlorpromazine 23-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 12788816-5 2003 Blockade of human ether-a-go-go-related gene (HERG) potassium channels, which plays a central role in arrhythmogenesis, has previously been reported to occur with chlorpromazine, but information on the mechanism of block is currently not available. Chlorpromazine 163-177 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 12788816-8 2003 (3) Chlorpromazine blocked HERG potassium channels with an IC(50) value of 21.6 micro M and a Hill coefficient of 1.11. Chlorpromazine 4-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 12788816-10 2003 (5) Inhibition of HERG channels by chlorpromazine displayed reverse frequency dependence, that is, the amount of block was lower at higher stimulation rates. Chlorpromazine 35-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 12759137-6 2003 Thus, as demonstrated for cardiac HERG channels in previous studies and for pancreatic beta-cell K(ATP) channels in the present study, fluoroquinolones differ markedly in their potencies to inhibit K(+) channel activity. Fluoroquinolones 135-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 12948624-0 2003 Phenytoin and phenobarbital inhibit human HERG potassium channels. Phenytoin 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 12948624-0 2003 Phenytoin and phenobarbital inhibit human HERG potassium channels. Phenobarbital 14-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 12948624-4 2003 Tail currents, which are purely related to HERG, were blocked with an IC50 (the concentration when 50% inhibition was obtained compared to control values) of 240 microM for PHT and 3 mM for PB. Phenobarbital 190-192 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 12626667-0 2003 Modulation of human ether-a-go-go-related K+ (HERG) channel inactivation by Cs+ and K+. Cesium 76-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 12695533-2 2003 Here we investigate the molecular mechanisms of voltage-dependent block of human ether-a-go-go-related gene (HERG) delayed rectifier K(+) channels expressed in Xenopus laevis oocytes by quinidine, an antiarrhythmic drug, and vesnarinone, a cardiotonic drug. Quinidine 186-195 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 12695533-2 2003 Here we investigate the molecular mechanisms of voltage-dependent block of human ether-a-go-go-related gene (HERG) delayed rectifier K(+) channels expressed in Xenopus laevis oocytes by quinidine, an antiarrhythmic drug, and vesnarinone, a cardiotonic drug. vesnarinone 225-236 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 12695533-4 2003 Block of HERG by quinidine (and its isomer quinine) was enhanced by progressive membrane depolarization and accompanied by a negative shift in the voltage dependence of channel activation. Quinidine 17-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 12695533-4 2003 Block of HERG by quinidine (and its isomer quinine) was enhanced by progressive membrane depolarization and accompanied by a negative shift in the voltage dependence of channel activation. Quinine 43-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 12695533-5 2003 As reported previously for other HERG blockers (e.g., MK-499, cisapride, terfenadine, chloroquine), the potency of quinidine was reduced >100-fold by the mutation of key aromatic residues (Y652, F656) located in the S6 domain. Quinidine 115-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 12695533-6 2003 Mutations of Y652 eliminated (Y652F) or reversed (Y652A) the voltage dependence of HERG channel block by quinidine and quinine. Quinidine 105-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 12695533-6 2003 Mutations of Y652 eliminated (Y652F) or reversed (Y652A) the voltage dependence of HERG channel block by quinidine and quinine. Quinine 119-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 12695533-8 2003 However, similar changes in the voltage-dependent profile for block of Y652F or Y652A HERG channels were observed with vesnarinone, a cardiotonic drug that is uncharged at physiological pH. vesnarinone 119-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 12775973-4 2003 In human ether-a-go-go (HERG)-transfected Chinese hamster ovary cells (n = 16), risperidone caused concentration-dependent block of the rapid component (I(Kr)) of the delayed rectifier potassium current with an IC(50) for tail block of 261 nM. Risperidone 80-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 12771193-0 2003 Saxitoxin is a gating modifier of HERG K+ channels. Saxitoxin 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 12771193-5 2003 STX decreased hERG K+ currents by stabilizing closed channel states visualized as shifts in the voltage dependence of channel opening to more depolarized membrane potentials. Saxitoxin 0-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 12771193-10 2003 The results are consistent with a simple model in which STX binds to the hERG K+ channel at multiple sites and alters the energetics of channel gating by shifting both the voltage-inactivation and voltage-activation processes. Saxitoxin 56-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 12771194-4 2003 Data from the other five functional mutants suggest that D411 can stabilize the HERG channel in the closed state, while D460 and D509 have the opposite effect. d411 57-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 12771194-6 2003 On the other hand, all five aspartates work in a concerted fashion in contributing to the slow deactivation process of the HERG channel. Aspartic Acid 28-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 123-127 12771194-7 2003 Accessibility tests of the introduced thiol groups to extracellular MTS reagents indicate that water-filled crevices penetrate deep into the HERG protein core, reaching the cytoplasmic halves of S1 and S2. Sulfhydryl Compounds 38-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 141-145 12771194-7 2003 Accessibility tests of the introduced thiol groups to extracellular MTS reagents indicate that water-filled crevices penetrate deep into the HERG protein core, reaching the cytoplasmic halves of S1 and S2. Water 95-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 141-145 12690509-2 2003 In this study, we have retrospectively screened specific founder mutations in KCNQ1 (KVLQT1) and KCNH2 (HERG) genes in 165 consecutive bodies found in water in Finland. Water 151-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-102 12690509-2 2003 In this study, we have retrospectively screened specific founder mutations in KCNQ1 (KVLQT1) and KCNH2 (HERG) genes in 165 consecutive bodies found in water in Finland. Water 151-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 12626667-1 2003 Unlike many other native and cloned K+ channels, human ether-a-go-go-related K+ (HERG) channels show significant Cs+ permeability with a PCs/PK (the permeability of Cs+ relative to that of K+) of 0.36 +/- 0.03 (n = 10). Cesium 113-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 12626667-1 2003 Unlike many other native and cloned K+ channels, human ether-a-go-go-related K+ (HERG) channels show significant Cs+ permeability with a PCs/PK (the permeability of Cs+ relative to that of K+) of 0.36 +/- 0.03 (n = 10). Cesium 165-168 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 12626667-2 2003 Here, we find that raising the concentration of external Cs+ (Cs+o) dramatically slows HERG channel inactivation without affecting activation. Cesium 57-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 12626667-8 2003 The strong effects of Cs+ on inactivation but not on activation highlight the importance of ion and channel interactions during the onset of inactivation in the HERG channel. Cesium 22-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 161-165 12650941-0 2003 Solution structure of CnErg1 (Ergtoxin), a HERG specific scorpion toxin. (6aR,9R)-N-[(1S,2S,4R,7S)-7-benzyl-2-hydroxy-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-5,8-dioxo-4,7-di(propan-2-yl)-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-7-(2-methylpropyl)-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide 30-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 12650941-1 2003 The three-dimensional structure of chemically synthesized CnErg1 (Ergtoxin), which specifically blocks HERG (human ether-a-go-go-related gene) K+ channels, was determined by nuclear magnetic resonance spectroscopy. (6aR,9R)-N-[(1S,2S,4R,7S)-7-benzyl-2-hydroxy-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-5,8-dioxo-4,7-di(propan-2-yl)-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-7-(2-methylpropyl)-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide 66-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 12618228-0 2003 Effects of propafenone and its main metabolite, 5-hydroxypropafenone, on HERG channels. Propafenone 11-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 12531891-4 2003 We found that both hyperglycemia (extracellular glucose concentration [Glu](o) = 10 or 20 mm) and hypoglycemia ([Glu](o) = 2.5, 1, or 0 mm) impaired HERG function by reducing HERG current (I(HERG)) density, as compared with normoglycemia ([Glu](o) = 5 mm). Glutamic Acid 71-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 12531891-4 2003 We found that both hyperglycemia (extracellular glucose concentration [Glu](o) = 10 or 20 mm) and hypoglycemia ([Glu](o) = 2.5, 1, or 0 mm) impaired HERG function by reducing HERG current (I(HERG)) density, as compared with normoglycemia ([Glu](o) = 5 mm). Glutamic Acid 113-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 12531891-4 2003 We found that both hyperglycemia (extracellular glucose concentration [Glu](o) = 10 or 20 mm) and hypoglycemia ([Glu](o) = 2.5, 1, or 0 mm) impaired HERG function by reducing HERG current (I(HERG)) density, as compared with normoglycemia ([Glu](o) = 5 mm). Glutamic Acid 113-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 12531891-9 2003 We conclude that ATP, derived from either glycolysis or oxidative phosphorylation, is critical for normal HERG function; depression of I(HERG) in hypoglycemia results from underproduction of ATP and in hyperglycemia from overproduction of reactive oxygen species. Adenosine Triphosphate 17-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 12531891-9 2003 We conclude that ATP, derived from either glycolysis or oxidative phosphorylation, is critical for normal HERG function; depression of I(HERG) in hypoglycemia results from underproduction of ATP and in hyperglycemia from overproduction of reactive oxygen species. Adenosine Triphosphate 17-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 12531891-9 2003 We conclude that ATP, derived from either glycolysis or oxidative phosphorylation, is critical for normal HERG function; depression of I(HERG) in hypoglycemia results from underproduction of ATP and in hyperglycemia from overproduction of reactive oxygen species. Adenosine Triphosphate 191-194 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 12531891-9 2003 We conclude that ATP, derived from either glycolysis or oxidative phosphorylation, is critical for normal HERG function; depression of I(HERG) in hypoglycemia results from underproduction of ATP and in hyperglycemia from overproduction of reactive oxygen species. Reactive Oxygen Species 239-262 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 12531891-3 2003 Here we studied modulation of human ether-a-go-go-related gene (HERG) K(+) channel, the major molecular component of delayed rectifier K(+) current responsible for cardiac repolarization, by glucose in HEK293 cells using whole-cell patch clamp techniques. Glucose 191-198 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 12531891-4 2003 We found that both hyperglycemia (extracellular glucose concentration [Glu](o) = 10 or 20 mm) and hypoglycemia ([Glu](o) = 2.5, 1, or 0 mm) impaired HERG function by reducing HERG current (I(HERG)) density, as compared with normoglycemia ([Glu](o) = 5 mm). Glucose 48-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 12593854-4 2003 The bulk of repolarization, including the after-hyperpolarization, was sustained by the human eag related (HERG) potassium current (I(HERG)) that also governs V(REST) in 21S cells. Potassium 113-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-111 12593854-4 2003 The bulk of repolarization, including the after-hyperpolarization, was sustained by the human eag related (HERG) potassium current (I(HERG)) that also governs V(REST) in 21S cells. Potassium 113-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 134-138 12618228-0 2003 Effects of propafenone and its main metabolite, 5-hydroxypropafenone, on HERG channels. 5-hydroxypropafenone 48-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 12618228-3 2003 In the present study we have analysed the effects of propafenone and 5-hydroxypropafenone on HERG current. Propafenone 53-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 12618228-3 2003 In the present study we have analysed the effects of propafenone and 5-hydroxypropafenone on HERG current. 5-hydroxypropafenone 69-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 12618228-9 2003 Propafenone, but not 5-hydroxypropafenone, inhibited to a higher extent HERG current at the end of 5-s depolarizing pulses to 0 mV than after promoting the transition of HERG channels from the inactivated to the opened state. Propafenone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 12618228-9 2003 Propafenone, but not 5-hydroxypropafenone, inhibited to a higher extent HERG current at the end of 5-s depolarizing pulses to 0 mV than after promoting the transition of HERG channels from the inactivated to the opened state. Propafenone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 170-174 12618228-10 2003 CONCLUSIONS: These results indicate that propafenone and its main active metabolite, 5-hydroxypropafenone, block HERG channels to a similar extent by binding predominantly to the open state of the channel. Propafenone 41-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 12618228-10 2003 CONCLUSIONS: These results indicate that propafenone and its main active metabolite, 5-hydroxypropafenone, block HERG channels to a similar extent by binding predominantly to the open state of the channel. 5-hydroxypropafenone 85-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 12689355-0 2003 Interaction between tetraethylammonium and permeant cations at the inactivation gate of the HERG potassium channel. Tetraethylammonium 20-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 12591761-2 2003 In this study, the effects of SP and its metabolite, canrenoic acid (CA), on human ether-a-go-go-related gene (HERG) currents were analyzed. Spironolactone 30-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 111-115 12591761-2 2003 In this study, the effects of SP and its metabolite, canrenoic acid (CA), on human ether-a-go-go-related gene (HERG) currents were analyzed. Canrenoic Acid 53-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 111-115 12591761-4 2003 SP decreased HERG currents in a concentration-dependent manner (IC50=23.0+/-1.5 micromol/L) and shifted the midpoint of the activation curve to more negative potentials (Vh=-13.1+/-3.4 versus -18.9+/-3.6 mV, P<0.05) without modifying the activation and deactivation kinetics. Spironolactone 0-2 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 12591761-11 2003 CONCLUSIONS: At concentrations reached after administration of therapeutic doses of SP, CA blocked the HERG channels by binding to both the closed and open states of the channel. Spironolactone 84-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 12689355-1 2003 The fast inactivation of the human ether-a-go-go related gene product (HERG) channel is a form of C-type inactivation and is decelerated by external tetraethylammonium (TEA) and potassium. Tetraethylammonium 149-167 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 12689355-1 2003 The fast inactivation of the human ether-a-go-go related gene product (HERG) channel is a form of C-type inactivation and is decelerated by external tetraethylammonium (TEA) and potassium. Tetraethylammonium 169-172 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 12689355-1 2003 The fast inactivation of the human ether-a-go-go related gene product (HERG) channel is a form of C-type inactivation and is decelerated by external tetraethylammonium (TEA) and potassium. Potassium 178-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 12634931-3 2003 The antiarrhythmic agent E-4031, a specific blocker of erg channels, served to isolate HERG currents as the drug-sensitive currents. E 4031 25-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 12538844-2 2003 Irbesartan exhibited a low affinity for HERG and KvLQT1+minK channels (IC(50) = 193.0 +/- 49.8 and 314.6 +/- 85.4 microM, respectively). Irbesartan 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 12538844-10 2003 Molecular modeling was used to define energy-minimized dockings of irbesartan to hKv1.5 and HERG channels. Irbesartan 67-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 12427763-6 2003 Sequence alignment against HERG showed a substitution of alanine for valine in the S4 domain. Alanine 57-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 12427763-6 2003 Sequence alignment against HERG showed a substitution of alanine for valine in the S4 domain. Valine 69-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 12427763-10 2003 Site-directed mutagenesis of alanine to valine in the S4 region of erg1-sm converted many of the properties to that of the cardiac HERG, including shifts in the voltage dependence of activation and slowing of deactivation. Alanine 29-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 12427763-10 2003 Site-directed mutagenesis of alanine to valine in the S4 region of erg1-sm converted many of the properties to that of the cardiac HERG, including shifts in the voltage dependence of activation and slowing of deactivation. Alanine 29-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 12427763-10 2003 Site-directed mutagenesis of alanine to valine in the S4 region of erg1-sm converted many of the properties to that of the cardiac HERG, including shifts in the voltage dependence of activation and slowing of deactivation. Valine 40-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 12427763-10 2003 Site-directed mutagenesis of alanine to valine in the S4 region of erg1-sm converted many of the properties to that of the cardiac HERG, including shifts in the voltage dependence of activation and slowing of deactivation. Valine 40-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 12524137-0 2003 Ethanol differently affects stress protein and HERG K+ channel expression in SH-SY5Y cells. Ethanol 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 12524137-4 2003 The GRP and human-ether-a-gogo-related gene (HERG) K(+)-channel expression were monitored in short- and long-term ethanol incubation experiments using the human neuroblastoma cell line SH-SY5Y. Ethanol 114-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 12522086-0 2003 Inhibition of hEAG1 and hERG1 potassium channels by clofilium and its tertiary analogue LY97241. clofilium 52-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-29 12522086-0 2003 Inhibition of hEAG1 and hERG1 potassium channels by clofilium and its tertiary analogue LY97241. LY 97241 88-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-29 12522086-2 2003 2 In the whole-cell mode of mammalian cells, LY97241 was shown to be a potent inhibitor of both hEAG1 and hERG1 channels (IC(50) of 4.9 and 2.2 nM, respectively). LY 97241 45-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-111 12361947-8 2002 Tyrosine phosphorylation of ERG-1 was decreased by transfection with SHP-1 wt and increased by SHP-1 Cys --> Ser. Tyrosine 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-33 12527373-2 2003 Wortmannin (a phosphoinositide 3-kinase (PI3K) inhibitor) caused approximately 30% reduction of HERG current (I(HERG)) stably expressed in HEK293 cells. Wortmannin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-100 12527373-2 2003 Wortmannin (a phosphoinositide 3-kinase (PI3K) inhibitor) caused approximately 30% reduction of HERG current (I(HERG)) stably expressed in HEK293 cells. Wortmannin 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-116 12528876-9 2003 CONCLUSIONS: Beat-to-beat nonalternating T-wave lability occurs in LQT1, LQT2, and LQT3 patients during catecholamine provocation and is associated with a history of prior cardiac events. Catecholamines 104-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 12361947-8 2002 Tyrosine phosphorylation of ERG-1 was decreased by transfection with SHP-1 wt and increased by SHP-1 Cys --> Ser. Cysteine 101-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-33 12361947-8 2002 Tyrosine phosphorylation of ERG-1 was decreased by transfection with SHP-1 wt and increased by SHP-1 Cys --> Ser. Serine 112-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-33 12460639-2 2002 We examined the detailed electrophysiological effects of nifekalant on human-ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes. nifekalant 57-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-103 12460639-2 2002 We examined the detailed electrophysiological effects of nifekalant on human-ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes. nifekalant 57-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 12460639-3 2002 Nifekalant inhibited the HERG current in a concentration-dependent manner with an IC(50) value of 7.9 microM although the drug did not inhibit the minK current in Xenopus oocytes, suggesting selective inhibition of the rapid component of the delayed rectifier K(+) current (I(Kr)) in cardiomyocytes. nifekalant 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 12460639-4 2002 Nifekalant showed a higher binding affinity for the open state than for the inactive state of HERG channels. nifekalant 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 12460639-5 2002 Nifekalant inhibited HERG channels in a frequency-dependent manner. nifekalant 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 12460639-7 2002 Nifekalant modified the voltage dependence and kinetics of HERG channel gating. nifekalant 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 12460639-8 2002 Thus, nifekalant inhibits HERG channels in a voltage-dependent and frequency-dependent manner, and the inhibitory effect may underlie the clinical efficacy of the drug against ventricular tachyarrhythmias. nifekalant 6-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 12411421-3 2002 The antihistamine terfenadine blocks HERG channels, and can cause QT prolongation and torsades de pointes, whereas its carboxylate fexofenadine lacks HERG blocking activity. antihistamine terfenadine 4-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 12354768-0 2002 A novel mutation (T65P) in the PAS domain of the human potassium channel HERG results in the long QT syndrome by trafficking deficiency. Aminosalicylic Acid 31-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 12354768-4 2002 We describe the identification and characterization of a novel missense mutation T65P in the PAS (Per-Arnt-Sim) domain of HERG, resulting in defective trafficking of the protein to the cell membrane. Aminosalicylic Acid 93-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 12354768-8 2002 This study is the first to relate a PAS domain mutation in HERG to a trafficking deficiency at body temperature, apart from effects on channel deactivation. Aminosalicylic Acid 36-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 12388652-2 2002 Our results show that LAAM, methadone, fentanyl, and buprenorphine were effective inhibitors of I(HERG), with IC(50) values in the 1 to 10 microM range. Methadyl Acetate 22-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 12388652-2 2002 Our results show that LAAM, methadone, fentanyl, and buprenorphine were effective inhibitors of I(HERG), with IC(50) values in the 1 to 10 microM range. Methadone 28-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 12388652-2 2002 Our results show that LAAM, methadone, fentanyl, and buprenorphine were effective inhibitors of I(HERG), with IC(50) values in the 1 to 10 microM range. Fentanyl 39-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 12388652-2 2002 Our results show that LAAM, methadone, fentanyl, and buprenorphine were effective inhibitors of I(HERG), with IC(50) values in the 1 to 10 microM range. Buprenorphine 53-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 12388652-4 2002 Compared with the reported maximal plasma concentration (C(max)) after administration of therapeutic doses of these drugs, the ratio of IC(50)/C(max) was highest for codeine and morphine (>455 and >400, respectively), thereby indicating that these drugs have the widest margin of safety (of the compounds tested) with respect to blockade of I(HERG). Codeine 166-173 potassium voltage-gated channel subfamily H member 2 Homo sapiens 349-353 12388652-4 2002 Compared with the reported maximal plasma concentration (C(max)) after administration of therapeutic doses of these drugs, the ratio of IC(50)/C(max) was highest for codeine and morphine (>455 and >400, respectively), thereby indicating that these drugs have the widest margin of safety (of the compounds tested) with respect to blockade of I(HERG). Morphine 178-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 349-353 12388652-6 2002 Further investigation showed that methadone block of I(HERG) was rapid, with steady-state inhibition achieved within 1 s when applied at its IC(50) concentration (10 microM) for I(HERG) block. Methadone 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 12388652-6 2002 Further investigation showed that methadone block of I(HERG) was rapid, with steady-state inhibition achieved within 1 s when applied at its IC(50) concentration (10 microM) for I(HERG) block. Methadone 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 12388652-9 2002 These results demonstrate that LAAM and methadone can block I(HERG) in transfected cells at clinically relevant concentrations, thereby providing a plausible mechanism for the adverse cardiac effects observed in some patients receiving LAAM or methadone. Methadyl Acetate 31-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 12388652-9 2002 These results demonstrate that LAAM and methadone can block I(HERG) in transfected cells at clinically relevant concentrations, thereby providing a plausible mechanism for the adverse cardiac effects observed in some patients receiving LAAM or methadone. Methadone 40-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 12388652-9 2002 These results demonstrate that LAAM and methadone can block I(HERG) in transfected cells at clinically relevant concentrations, thereby providing a plausible mechanism for the adverse cardiac effects observed in some patients receiving LAAM or methadone. Methadyl Acetate 236-240 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 12388652-9 2002 These results demonstrate that LAAM and methadone can block I(HERG) in transfected cells at clinically relevant concentrations, thereby providing a plausible mechanism for the adverse cardiac effects observed in some patients receiving LAAM or methadone. Methadone 244-253 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 12359267-0 2002 Fenamate-induced enhancement of heterologously expressed HERG currents in Xenopus oocytes. Fenamates 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 12359267-3 2002 In this study, we examined the effects of the non-steroidal anti-inflammatory agents, flufenamic acid and niflumic acid, on heterologously expressed HERG channels in oocytes. Flufenamic Acid 86-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 12359267-3 2002 In this study, we examined the effects of the non-steroidal anti-inflammatory agents, flufenamic acid and niflumic acid, on heterologously expressed HERG channels in oocytes. Niflumic Acid 106-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 12359267-7 2002 Fenamates accelerated the activation rate of HERG channels and decelerated their deactivation. Fenamates 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 12359267-11 2002 The effects of the fenamates were blocked by the HERG channel blocker, E-4031 and were also not observed in water-injected oocytes. Fenamates 19-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 12359267-11 2002 The effects of the fenamates were blocked by the HERG channel blocker, E-4031 and were also not observed in water-injected oocytes. E 4031 71-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 12359267-12 2002 Our data suggest that fenamates enhance HERG currents and affect the action potential duration in the heart. Fenamates 22-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 12411421-5 2002 In the present study the ability of fexofenadine to block the K897T HERG channel variant was investigated. fexofenadine 36-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 12431311-7 2002 In this review article, sympathetic stimulation with isoproterenol or epinephrine infusion is demonstrated to modulate differentially these repolarization indices in the ECG as well as the action potentials of the three cells between the LQT1, LQT2, and LQT3 syndromes both experimentally and clinically, explaining the differences in the sensitivity of genotypes of congenital LQTS to sympathetic stimulation. Isoproterenol 53-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 244-248 12431311-7 2002 In this review article, sympathetic stimulation with isoproterenol or epinephrine infusion is demonstrated to modulate differentially these repolarization indices in the ECG as well as the action potentials of the three cells between the LQT1, LQT2, and LQT3 syndromes both experimentally and clinically, explaining the differences in the sensitivity of genotypes of congenital LQTS to sympathetic stimulation. Epinephrine 70-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 244-248 12176106-7 2002 We found that sertindole, pimozide and thioridazine displayed little (<10-fold) or no selectivity for dopamine D(2) or 5-HT(2A) receptors relative to their HERG channel affinities. Thioridazine 39-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 159-163 12176129-0 2002 The anti-malarial drug halofantrine and its metabolite N-desbutylhalofantrine block HERG potassium channels. halofantrine 23-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 12176129-0 2002 The anti-malarial drug halofantrine and its metabolite N-desbutylhalofantrine block HERG potassium channels. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 55-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 12176129-3 2002 METHODS: We studied the effects of halofantrine (0.1-1000 nM) and its major metabolite N-desbutylhalofantrine (3-1000 nM) on wild type HERG K(+) channels stably expressed in HEK 293 cells, using the whole cell patch-clamp recording technique. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 87-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 135-139 12176129-4 2002 RESULTS: Halofantrine and N-desbutylhalofantrine blocked HERG K(+) channels in a concentration-dependent manner with a half-maximal inhibitory concentration of 21.6 nM (n=31 cells) and 71.7 nM (n=18 cells), respectively. halofantrine 9-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 12176129-4 2002 RESULTS: Halofantrine and N-desbutylhalofantrine blocked HERG K(+) channels in a concentration-dependent manner with a half-maximal inhibitory concentration of 21.6 nM (n=31 cells) and 71.7 nM (n=18 cells), respectively. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 26-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 12176129-7 2002 Using a ventricular action potential voltage clamp protocol, halofantrine and N-desbutylhalofantrine block of HERG current was greatest during phases 2 and 3 of the action potential waveform. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 78-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 12176129-8 2002 CONCLUSION: We conclude that both halofantrine and N-desbutylhalofantrine cause high affinity block of HERG K(+) channels. halofantrine 34-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 12176129-8 2002 CONCLUSION: We conclude that both halofantrine and N-desbutylhalofantrine cause high affinity block of HERG K(+) channels. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 51-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 12176106-9 2002 Of the other drugs tested (ziprasidone, quetiapine, risperidone and olanzapine), olanzapine displayed the greatest selectivity for dopamine D(2) and 5-HT(2A) receptor binding (100-1000-fold) compared to its HERG channel IC(50). Olanzapine 81-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 207-211 12176106-9 2002 Of the other drugs tested (ziprasidone, quetiapine, risperidone and olanzapine), olanzapine displayed the greatest selectivity for dopamine D(2) and 5-HT(2A) receptor binding (100-1000-fold) compared to its HERG channel IC(50). dopamine d 131-141 potassium voltage-gated channel subfamily H member 2 Homo sapiens 207-211 12357160-0 2002 Kinetic modulation of HERG potassium channels by the volatile anesthetic halothane. Halothane 73-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 12357160-2 2002 General anesthetics, like halothane, can prolong Q-T interval, suggesting that they act on myocellular repolarization, possibly involving HERG channels. Halothane 26-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 138-142 12357160-3 2002 Evidence for direct modulation of HERG channels by halothane is still lacking. Halothane 51-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 12357160-4 2002 To gain insight on HERG channel modulation by halothane the authors recorded macroscopic currents expressed in Xenopus oocytes and conducted non-stationary noise analysis to evaluate single channel parameters modified by the anesthetic. Halothane 46-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 12357160-12 2002 CONCLUSIONS: Halothane inhibits HERG currents expressed in oocytes in a concentration-dependent manner. Halothane 13-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 12357160-14 2002 The authors" results demonstrate that halothane decreased HERG currents by modulating kinetic properties of HERG channels, decreasing their open probability. Halothane 38-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 12357160-14 2002 The authors" results demonstrate that halothane decreased HERG currents by modulating kinetic properties of HERG channels, decreasing their open probability. Halothane 38-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 108-112 12356854-0 2002 Extracellular sodium interacts with the HERG channel at an outer pore site. Sodium 14-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 12208728-4 2002 Here, we show that HERG conductance markedly promotes H2O2-induced apoptosis of various tumor cells, whereas HERG expression facilitates the tumor cell proliferation caused by tumor necrosis factor (TNF) ligand (TNF-alpha). Hydrogen Peroxide 54-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 12065733-0 2002 Blockade of human cardiac potassium channel human ether-a-go-go-related gene (HERG) by macrolide antibiotics. macrolide antibiotics 87-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 12021266-4 2002 Streptavidin selection of biotin-labeled surface proteins showed good expression of wild-type and HERG(Delta159) at the cell surface and low expression of HERG(Delta147-LGL) and HERG(Delta147). Biotin 26-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 12021266-4 2002 Streptavidin selection of biotin-labeled surface proteins showed good expression of wild-type and HERG(Delta159) at the cell surface and low expression of HERG(Delta147-LGL) and HERG(Delta147). Biotin 26-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 168-172 12021266-4 2002 Streptavidin selection of biotin-labeled surface proteins showed good expression of wild-type and HERG(Delta159) at the cell surface and low expression of HERG(Delta147-LGL) and HERG(Delta147). Biotin 26-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 12142119-5 2002 Deoxyribonucleic acid samples were genotyped for the nucleotide 2690A>C variation of the HERG gene, corresponding to the HERG K(lysine)897T(threonine) amino acid polymorphism. Lysine 131-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 12142119-5 2002 Deoxyribonucleic acid samples were genotyped for the nucleotide 2690A>C variation of the HERG gene, corresponding to the HERG K(lysine)897T(threonine) amino acid polymorphism. Lysine 131-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 12142119-5 2002 Deoxyribonucleic acid samples were genotyped for the nucleotide 2690A>C variation of the HERG gene, corresponding to the HERG K(lysine)897T(threonine) amino acid polymorphism. Threonine 143-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 12142119-5 2002 Deoxyribonucleic acid samples were genotyped for the nucleotide 2690A>C variation of the HERG gene, corresponding to the HERG K(lysine)897T(threonine) amino acid polymorphism. Threonine 143-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 12142125-9 2002 Nicorandil caused greater shortening of ARI and greater attenuation of transmural ARI dispersion in the LQT2 model than in the LQT3 model. Nicorandil 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 12142125-10 2002 After treatment with nicorandil, a single VTA was induced in the LQT2 model by LSS, whereas in the LQT3 model, VTA remained inducible by BRADY in four experiments and LSS in one experiment. Nicorandil 21-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 12142125-12 2002 Nicorandil attenuated the heterogeneity of ventricular repolarization and suppressed the induction of VTA in the LQT2 model, but had a limited therapeutic effect in the LQT3 model. Nicorandil 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 12063283-6 2002 Cardiac action potential duration was prolonged by antagonists of either ERG1 (MK-499, cisapride) or KCNQ1/KCNE1 (chromanol 293B). L 706000 79-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 12086981-0 2002 Inhibition of the current of heterologously expressed HERG potassium channels by flecainide and comparison with quinidine, propafenone and lignocaine. Flecainide 81-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 12086981-3 2002 In this study, we investigated the effects of the Class Ic antiarrhythmic agent flecainide (FLEC) on ionic current (I(HERG)) mediated by cloned HERG channels at 37 degrees C. We also compared the inhibitory potency of FLEC with other Class I agents: quinidine (QUIN, Class Ia); lignocaine (LIG, Class Ib) and propafenone (PROPAF, Class Ic). Flecainide 80-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-122 12086981-3 2002 In this study, we investigated the effects of the Class Ic antiarrhythmic agent flecainide (FLEC) on ionic current (I(HERG)) mediated by cloned HERG channels at 37 degrees C. We also compared the inhibitory potency of FLEC with other Class I agents: quinidine (QUIN, Class Ia); lignocaine (LIG, Class Ib) and propafenone (PROPAF, Class Ic). Flecainide 80-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 144-148 12086981-3 2002 In this study, we investigated the effects of the Class Ic antiarrhythmic agent flecainide (FLEC) on ionic current (I(HERG)) mediated by cloned HERG channels at 37 degrees C. We also compared the inhibitory potency of FLEC with other Class I agents: quinidine (QUIN, Class Ia); lignocaine (LIG, Class Ib) and propafenone (PROPAF, Class Ic). Flecainide 92-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-122 12086981-3 2002 In this study, we investigated the effects of the Class Ic antiarrhythmic agent flecainide (FLEC) on ionic current (I(HERG)) mediated by cloned HERG channels at 37 degrees C. We also compared the inhibitory potency of FLEC with other Class I agents: quinidine (QUIN, Class Ia); lignocaine (LIG, Class Ib) and propafenone (PROPAF, Class Ic). Flecainide 92-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 144-148 12086981-15 2002 Under similar recording conditions QUIN inhibited I(HERG) with an IC(50) of 0.41+/-0.04 microM and PROPAF inhibited I(HERG) with an IC(50) of 0.44+/-0.07 microM. Quinidine 35-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 12086981-21 2002 The rank potency as HERG blockers of the Class I drugs tested in this study was QUIN=PROPAF>FLEC>>LIG. Quinidine 80-84 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 12065733-2 2002 To clarify the underlying ionic mechanisms, we examined the effects of six macrolides on the human ether-a-go-go-related gene (HERG)-encoded potassium current stably expressed in human embryonic kidney-293 cells. Potassium 141-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 12065733-6 2002 Mechanistic studies showed that inhibition of HERG current by clarithromycin did not require activation of the channel and was both voltage- and time-dependent. Clarithromycin 62-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 12070109-5 2002 We tested terfenadine carboxylate (fexofenadine) and terfenadine, structurally similar drugs with markedly different affinities for HERG block, for rescue of trafficking-defective LQT2 mutations. fexofenadine 10-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 12084597-6 2002 RESULTS: Propranolol in the absence of epinephrine significantly prolonged the mean cQT(p) value but not the mean cQT(e) value, thus decreasing the mean cT(p-e) value in both LQT1 and LQT2 patients; the differences with propranolol were significantly larger in LQT1 than in LQT2 (p < 0.05). Propranolol 9-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 184-188 11960982-2 2002 Here we investigate the voltage-dependent block of wild-type and mutant human ether-a-go-go related gene (HERG) K(+) channels by the antimalarial compound chloroquine. Chloroquine 155-166 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 11960982-5 2002 Chloroquine also slowed the apparent rate of HERG deactivation, reflecting the inability of drug-bound channels to close. Chloroquine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 11960982-6 2002 Mutation to alanine of aromatic residues (Tyr-652 or Phe-656) located in the S6 domain of HERG greatly reduced the potency of channel block by chloroquine (IC(50) > 1 mm at 0 mV). Alanine 12-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 11960982-6 2002 Mutation to alanine of aromatic residues (Tyr-652 or Phe-656) located in the S6 domain of HERG greatly reduced the potency of channel block by chloroquine (IC(50) > 1 mm at 0 mV). Tyrosine 42-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 11960982-6 2002 Mutation to alanine of aromatic residues (Tyr-652 or Phe-656) located in the S6 domain of HERG greatly reduced the potency of channel block by chloroquine (IC(50) > 1 mm at 0 mV). Phenylalanine 53-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 11960982-6 2002 Mutation to alanine of aromatic residues (Tyr-652 or Phe-656) located in the S6 domain of HERG greatly reduced the potency of channel block by chloroquine (IC(50) > 1 mm at 0 mV). Chloroquine 143-154 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 11960982-9 2002 HERG channel block was voltage-independent when the hydroxyl group of Tyr-652 was removed by mutating the residue to Phe. Tyrosine 70-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11960982-9 2002 HERG channel block was voltage-independent when the hydroxyl group of Tyr-652 was removed by mutating the residue to Phe. Phenylalanine 117-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11960982-10 2002 Together these findings indicate a critical role for Tyr-652 in voltage-dependent block of HERG channels. Tyrosine 53-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11960982-11 2002 Molecular modeling was used to define energy-minimized dockings of chloroquine to the central cavity of HERG. Chloroquine 67-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 11960982-12 2002 Our experimental findings and modeling suggest that chloroquine preferentially blocks open HERG channels by cation-pi and pi-stacking interactions with Tyr-652 and Phe-656 of multiple subunits. Chloroquine 52-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11893742-7 2002 The selective HERG channel blocker, E-4031, reduced proliferation of CEM, U937, and K562 cells, and this appears to be the first direct evidence of a functional role for the HERG current in cancer cells. E 4031 36-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 11960982-12 2002 Our experimental findings and modeling suggest that chloroquine preferentially blocks open HERG channels by cation-pi and pi-stacking interactions with Tyr-652 and Phe-656 of multiple subunits. Tyrosine 152-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11960982-12 2002 Our experimental findings and modeling suggest that chloroquine preferentially blocks open HERG channels by cation-pi and pi-stacking interactions with Tyr-652 and Phe-656 of multiple subunits. Phenylalanine 164-167 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 12029369-7 2002 It also demonstrates that both proximal and eag/PAS domains in the amino terminus contribute to set the gating characteristics of HERG channels. Protactinium 48-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 12069453-7 2002 Epinephrine also prolonged the QTc dramatically (502+/-23 to 620+/-39 ms; P<0.0005, +24%) at peak of epinephrine in LQT2 patients, but this shortened to baseline levels at steady state (531+/-25 ms; P=ns vs baseline, +6%). Epinephrine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 119-123 12069453-7 2002 Epinephrine also prolonged the QTc dramatically (502+/-23 to 620+/-39 ms; P<0.0005, +24%) at peak of epinephrine in LQT2 patients, but this shortened to baseline levels at steady state (531+/-25 ms; P=ns vs baseline, +6%). Epinephrine 104-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 119-123 11864984-2 2002 Potential insight into the coupling mechanism was provided by our previous finding that mutation to Lys of a single residue (Asp(540)) located in the S4-S5 linker endowed HERG (human ether-a-go-go-related gene) K(+) channels with the unusual ability to open in response to membrane depolarization and hyperpolarization in a voltage-dependent manner. Lysine 100-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 11864984-2 2002 Potential insight into the coupling mechanism was provided by our previous finding that mutation to Lys of a single residue (Asp(540)) located in the S4-S5 linker endowed HERG (human ether-a-go-go-related gene) K(+) channels with the unusual ability to open in response to membrane depolarization and hyperpolarization in a voltage-dependent manner. Aspartic Acid 125-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 11864984-4 2002 Therefore, we mutated six residues located in this region of S6 (Ile(662)-Tyr(667)) to Ala in D540K HERG channels. Isoleucine 65-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 11864984-5 2002 Mutation of Arg(665), but not the other five residues, prevented hyperpolarization-dependent reopening of D540K HERG channels. Arginine 12-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-116 11864984-8 2002 Together these findings suggest that a single residue (Arg(665)) in the S6 domain interacts with Lys(540) by electrostatic repulsion to couple voltage sensing to hyperpolarization-dependent opening of D540K HERG K(+) channels. Arginine 55-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 207-211 11864984-8 2002 Together these findings suggest that a single residue (Arg(665)) in the S6 domain interacts with Lys(540) by electrostatic repulsion to couple voltage sensing to hyperpolarization-dependent opening of D540K HERG K(+) channels. Lysine 97-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 207-211 11893742-7 2002 The selective HERG channel blocker, E-4031, reduced proliferation of CEM, U937, and K562 cells, and this appears to be the first direct evidence of a functional role for the HERG current in cancer cells. E 4031 36-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 12133532-4 2002 RESULTS: Elevation of the intracellular cAMP-concentration by incubation with the adenylate cyclase activator, forskolin (10 micromol/L), and the broad range phosphodiesterase inhibitor, IBMX (100 micromol/L), caused a HERG tail current reduction of 83.2%. Cyclic AMP 40-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 219-223 11864985-2 2002 We apply cysteine-scanning mutagenesis to the S5-P and P-S6 linkers of HERG and examine the resulting changes in ErgTx potency. Cysteine 9-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 71-75 12133532-9 2002 The coupling between the repolarizing cardiac HERG potassium current and the protein kinase A system could contribute to arrhythmogenesis under pathophysiological conditions. Potassium 51-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 12062363-3 2002 Therefore, we investigated the role of a glutamic acid at the vicinity of the pore in HERG channels by mutating it to a lysine. Glutamic Acid 41-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 11834728-4 2002 In this first report of regulation by tyrosine phosphorylation we show that MLS-9 cells express transcripts for r-erg1 (rat homologue of HERG) and r-erg2, and an immunoreactive doublet was identified using an anti-HERG antibody. Tyrosine 38-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 11834728-4 2002 In this first report of regulation by tyrosine phosphorylation we show that MLS-9 cells express transcripts for r-erg1 (rat homologue of HERG) and r-erg2, and an immunoreactive doublet was identified using an anti-HERG antibody. Tyrosine 38-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 214-218 11953308-3 2002 Stress stimulates beta-adrenergic receptors, leading to cAMP elevations that can regulate HERG K+ channels both directly and via phosphorylation by cAMP-dependent protein kinase (PKA). Cyclic AMP 56-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 11953308-4 2002 We show that HERG associates with 14-3-3epsilon to potentiate cAMP/PKA effects upon HERG. Cyclic AMP 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 11953308-4 2002 We show that HERG associates with 14-3-3epsilon to potentiate cAMP/PKA effects upon HERG. Cyclic AMP 62-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 11927665-7 2002 There were major discrepancies between the sensitivity to quinidine, E-4031 and dofetilide of I(HERG) in Xenopus oocytes compared to I(Kr), which were not substantially affected by coexpression with MiRP1. dofetilide 80-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-101 11756457-1 2002 The K(+) channels encoded by the human Ether-a-gogo Related Gene-1 (hERG1) are crucially involved in controlling heart and brain excitability and are selectively influenced by reactive oxygen species (ROS). Reactive Oxygen Species 176-199 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-73 11756457-1 2002 The K(+) channels encoded by the human Ether-a-gogo Related Gene-1 (hERG1) are crucially involved in controlling heart and brain excitability and are selectively influenced by reactive oxygen species (ROS). Reactive Oxygen Species 201-204 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-73 11756457-2 2002 To localize the molecular regions involved in ROS-induced modulation of hERG1, segmental exchanges between the ROS-sensitive hERG1 and the ROS-insensitive bovine ether-a-gogo gene (bEAG) K(+) channels were generated, and the sensitivity of these chimeric channels to ROS was studied with the two-microelectrode voltage-clamp technique upon their expression in Xenopus oocytes. Reactive Oxygen Species 46-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 11756457-2 2002 To localize the molecular regions involved in ROS-induced modulation of hERG1, segmental exchanges between the ROS-sensitive hERG1 and the ROS-insensitive bovine ether-a-gogo gene (bEAG) K(+) channels were generated, and the sensitivity of these chimeric channels to ROS was studied with the two-microelectrode voltage-clamp technique upon their expression in Xenopus oocytes. Reactive Oxygen Species 111-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-130 11756457-2 2002 To localize the molecular regions involved in ROS-induced modulation of hERG1, segmental exchanges between the ROS-sensitive hERG1 and the ROS-insensitive bovine ether-a-gogo gene (bEAG) K(+) channels were generated, and the sensitivity of these chimeric channels to ROS was studied with the two-microelectrode voltage-clamp technique upon their expression in Xenopus oocytes. Reactive Oxygen Species 111-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-130 11756457-2 2002 To localize the molecular regions involved in ROS-induced modulation of hERG1, segmental exchanges between the ROS-sensitive hERG1 and the ROS-insensitive bovine ether-a-gogo gene (bEAG) K(+) channels were generated, and the sensitivity of these chimeric channels to ROS was studied with the two-microelectrode voltage-clamp technique upon their expression in Xenopus oocytes. Reactive Oxygen Species 111-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-130 11756457-3 2002 Substitution of the S(5)-S(6) linker of hERG1 with the corresponding bEAG region removed channel sensitivity to ROS, whereas the reverse chimeric exchange introduced ROS sensitivity into bEAG. Reactive Oxygen Species 112-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-45 11756457-4 2002 Mutation of each of the two hERG1 histidines at positions 578 and 587 within the S(5)-S(6) linker generated K(+) channels insensitive to modulation by ROS. Histidine 34-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-33 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). Iron 21-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-118 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). Iron 21-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-160 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). Deferoxamine 36-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-118 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). Deferoxamine 36-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-160 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). 1,10-phenanthroline 63-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-118 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). 1,10-phenanthroline 63-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-160 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). Reactive Oxygen Species 187-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-118 11756457-5 2002 In addition, the two iron chelators desferrioxamine (1 mm) and o-phenanthroline (0.2 mm) significantly inhibited hERG1 outward K(+) currents and prevented hERG1 inhibition induced by the ROS-scavenging enzyme catalase (1000 units/ml). Reactive Oxygen Species 187-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-160 11756457-6 2002 Finally, the hERG1-inhibitory effect exerted by the iron chelators was prevented by the hERG1 H578D/H587Y double mutation. Iron 52-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-18 11756457-6 2002 Finally, the hERG1-inhibitory effect exerted by the iron chelators was prevented by the hERG1 H578D/H587Y double mutation. Iron 52-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-93 11756457-7 2002 Collectively, the results obtained suggest that histidines at positions 578 and 587 in the S(5)-S(6) linker region of hERG1 K(+) channels are crucial players in ROS-induced modulation of hERG1 K(+) channels. Histidine 48-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-123 11756457-7 2002 Collectively, the results obtained suggest that histidines at positions 578 and 587 in the S(5)-S(6) linker region of hERG1 K(+) channels are crucial players in ROS-induced modulation of hERG1 K(+) channels. Histidine 48-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 187-192 11756457-7 2002 Collectively, the results obtained suggest that histidines at positions 578 and 587 in the S(5)-S(6) linker region of hERG1 K(+) channels are crucial players in ROS-induced modulation of hERG1 K(+) channels. Reactive Oxygen Species 161-164 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-123 11756457-7 2002 Collectively, the results obtained suggest that histidines at positions 578 and 587 in the S(5)-S(6) linker region of hERG1 K(+) channels are crucial players in ROS-induced modulation of hERG1 K(+) channels. Reactive Oxygen Species 161-164 potassium voltage-gated channel subfamily H member 2 Homo sapiens 187-192 12645305-29 2002 TRH modifies the current kinetics of human HERG potassium channel co-expressed in Xenopus oocytes with the TRH receptor, whose activity is modulated via the protein kinase C pathway linked to a G protein-coupled receptor and is regulated by changes in the PIP2 concentration in the membrane. Phosphatidylinositol 4,5-Diphosphate 256-260 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 11852052-0 2002 Inhibitory actions of the selective serotonin re-uptake inhibitor citalopram on HERG and ventricular L-type calcium currents. Serotonin 36-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 11852052-0 2002 Inhibitory actions of the selective serotonin re-uptake inhibitor citalopram on HERG and ventricular L-type calcium currents. Citalopram 66-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 11852052-1 2002 Using whole-cell patch clamp recording of heterologous HERG-mediated currents in transfected mammalian cells, we observed that the selective serotonin re-uptake inhibitor citalopram blocks HERG with an IC(50) of 3.97 microM. Serotonin 141-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 11852052-1 2002 Using whole-cell patch clamp recording of heterologous HERG-mediated currents in transfected mammalian cells, we observed that the selective serotonin re-uptake inhibitor citalopram blocks HERG with an IC(50) of 3.97 microM. Serotonin 141-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 189-193 11852052-1 2002 Using whole-cell patch clamp recording of heterologous HERG-mediated currents in transfected mammalian cells, we observed that the selective serotonin re-uptake inhibitor citalopram blocks HERG with an IC(50) of 3.97 microM. Citalopram 171-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 11852052-1 2002 Using whole-cell patch clamp recording of heterologous HERG-mediated currents in transfected mammalian cells, we observed that the selective serotonin re-uptake inhibitor citalopram blocks HERG with an IC(50) of 3.97 microM. Citalopram 171-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 189-193 11852052-6 2002 The effects of citalopram on both calcium current amplitude and the I(Ca,L) "window" may help to explain citalopram"s good cardiac safety profile, given its propensity to block HERG at excessive dosages. Citalopram 15-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 177-181 11852052-6 2002 The effects of citalopram on both calcium current amplitude and the I(Ca,L) "window" may help to explain citalopram"s good cardiac safety profile, given its propensity to block HERG at excessive dosages. Citalopram 105-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 177-181 11805215-0 2002 The antidepressant drug fluoxetine is an inhibitor of human ether-a-go-go-related gene (HERG) potassium channels. Fluoxetine 24-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 11805215-5 2002 We found that fluoxetine blocked HERG channels with an IC(50) value of 3.1 microM. Fluoxetine 14-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 11805215-9 2002 This is the first study demonstrating that HERG potassium channels are blocked by the selective serotonin reuptake inhibitor fluoxetine. Fluoxetine 125-135 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 11755529-1 2002 We show here that ergtoxin (ErgTx) is a bona fide, specific blocker of the human ether-a-go-go-related gene (HERG) channels. (6aR,9R)-N-[(1S,2S,4R,7S)-7-benzyl-2-hydroxy-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-5,8-dioxo-4,7-di(propan-2-yl)-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide;(6aR,9R)-N-[(1S,2S,4R,7S)-2-hydroxy-7-(2-methylpropyl)-5,8-dioxo-4-propan-2-yl-3-oxa-6,9-diazatricyclo[7.3.0.02,6]dodecan-4-yl]-7-methyl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide 18-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 11755529-4 2002 From the P-region point mutants of HERG channel assays, only the mutant N598Q shows about 25% decrement of the ErgTx inhibitory effect. n598q 72-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 11744013-0 2002 Inhibition of HERG potassium channels by cocaethylene: a metabolite of cocaine and ethanol. cocaethylene 41-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 11744013-0 2002 Inhibition of HERG potassium channels by cocaethylene: a metabolite of cocaine and ethanol. Cocaine 71-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 11744013-0 2002 Inhibition of HERG potassium channels by cocaethylene: a metabolite of cocaine and ethanol. Ethanol 83-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 11744013-3 2002 RESULTS: The cocaethylene inhibition of HERG is concentration-dependent with an IC(50) of 4.0 microM. cocaethylene 13-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 11744013-7 2002 CONCLUSIONS: Cocaethylene inhibits HERG by binding to the activated or open channels and by modulating the kinetics of inactivation. cocaethylene 13-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 11926362-1 2002 In this paper, we describe an assay using radioactive rubidium (86Rb) efflux to screen functional human ether-a go-go-related gene (HERG) K+ channels in a high-throughput screening (HTS) format. Rubidium 54-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-136 11926362-1 2002 In this paper, we describe an assay using radioactive rubidium (86Rb) efflux to screen functional human ether-a go-go-related gene (HERG) K+ channels in a high-throughput screening (HTS) format. Rubidium-86 64-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-136 11926362-5 2002 Our results provide some explanations for the variances of the assay results and offer some guidelines for using the Rb efflux assay to evaluate compound interactions with HERG K+ channels in the pharmaceutical industry. Rubidium 117-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 172-176 11994029-9 2002 Verapamil is a notable example of a false positive: it blocks human ether-a-go-go-related (HERG) K(+) channels, but is reported to have little potential to trigger torsade de pointes. Verapamil 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11862331-3 2002 In oocytes using the two electrode voltage clamp technique potassium currents of hminK-, HERG- and Kir2.2-expressing oocytes were inhibited by pentobarbital with IC50 values of 0.20, 1.58 and 0.54 mM, respectively. Potassium 59-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 11862331-3 2002 In oocytes using the two electrode voltage clamp technique potassium currents of hminK-, HERG- and Kir2.2-expressing oocytes were inhibited by pentobarbital with IC50 values of 0.20, 1.58 and 0.54 mM, respectively. Pentobarbital 143-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 11862331-5 2002 Pentobarbital-induced HERG inhibition was not dependent on voltage but influenced the deactivation kinetics and shifted half-maximal activation to more negative voltages. Pentobarbital 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 11714889-3 2001 Cisapride is a potent blocker of human ether-a-gogo (HERG) K(+) channels and prolongs the cardiac action potential in a reverse use dependence manner. Cisapride 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 53-57 11739282-0 2001 HERG K(+) channel activity is regulated by changes in phosphatidyl inositol 4,5-bisphosphate. Phosphatidylinositol 4,5-Diphosphate 54-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11739282-3 2001 We sought to investigate whether PIP2 changes could alter HERG K(+) channel activity in a manner similar to that seen with inward rectifier channels. Phosphatidylinositol 4,5-Diphosphate 33-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 11739282-7 2001 PIP2 significantly attenuated the run-down of HERG channel activity that we normally observe after patch excision, suggesting that channel run-down is due, in part, to membrane depletion of PIP2. Phosphatidylinositol 4,5-Diphosphate 0-4 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 11739282-7 2001 PIP2 significantly attenuated the run-down of HERG channel activity that we normally observe after patch excision, suggesting that channel run-down is due, in part, to membrane depletion of PIP2. Phosphatidylinositol 4,5-Diphosphate 190-194 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 11739282-9 2001 The physiological relevance of PIP2-HERG interactions is supported by our finding that phenylephrine reduced the K(+) current density in cells coexpressing alpha1A-receptor and HERG. Phosphatidylinositol 4,5-Diphosphate 31-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 11739282-9 2001 The physiological relevance of PIP2-HERG interactions is supported by our finding that phenylephrine reduced the K(+) current density in cells coexpressing alpha1A-receptor and HERG. Phosphatidylinositol 4,5-Diphosphate 31-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 177-181 11739282-9 2001 The physiological relevance of PIP2-HERG interactions is supported by our finding that phenylephrine reduced the K(+) current density in cells coexpressing alpha1A-receptor and HERG. Phenylephrine 87-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 11739282-9 2001 The physiological relevance of PIP2-HERG interactions is supported by our finding that phenylephrine reduced the K(+) current density in cells coexpressing alpha1A-receptor and HERG. Phenylephrine 87-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 177-181 11739282-11 2001 Thus, dynamic regulation of HERG K(+) channels may be achieved via receptor-mediated changes in PIP2 concentrations. Phosphatidylinositol 4,5-Diphosphate 96-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-32 11723241-2 2001 HERG/I(Kr) channels are blocked selectively by class III antiarrhythmic methanesulfonanilide drugs such as dofetilide. dofetilide 107-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11723241-12 2001 We conclude that high affinity methanesulfonanilide binding to HERG channels is strongly dependent on C-type inactivation. N-Phenylmethanesulfonamide 31-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 11723012-0 2001 Phospholipid metabolite 1-palmitoyl-lysophosphatidylcholine enhances human ether-a-go-go-related gene (HERG) K(+) channel function. Phospholipids 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 11723012-0 2001 Phospholipid metabolite 1-palmitoyl-lysophosphatidylcholine enhances human ether-a-go-go-related gene (HERG) K(+) channel function. We 201 24-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 11736694-8 2001 E-4031, a blocker of a novel inwardly rectifying K+ channel, i.e. the human ether-a-go-go-related gene (HERG) K+ channel, significantly increased contraction amplitude. E 4031 0-6 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 11802537-5 2001 Genotyping revealed a novel nonsense mutation, R744X (C to T transition in codon 744), in the KCNH2 potassium channel gene, resulting in truncation of the putative cyclic nucleotide-binding domain and C-terminal region of the HERG K(+)-channel in all affected family members. Nucleotides, Cyclic 164-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-99 11802537-5 2001 Genotyping revealed a novel nonsense mutation, R744X (C to T transition in codon 744), in the KCNH2 potassium channel gene, resulting in truncation of the putative cyclic nucleotide-binding domain and C-terminal region of the HERG K(+)-channel in all affected family members. Nucleotides, Cyclic 164-181 potassium voltage-gated channel subfamily H member 2 Homo sapiens 226-230 11698075-0 2001 [3H]dofetilide binding to HERG transfected membranes: a potential high throughput preclinical screen. Tritium 1-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 11698075-0 2001 [3H]dofetilide binding to HERG transfected membranes: a potential high throughput preclinical screen. dofetilide 4-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 11698075-1 2001 The pharmacological characteristics of [3H]dofetilide binding were examined in membranes prepared from human embryonic kidney (HEK293) cells stably expressing human ether-a-go-go related gene (HERG) K+ channels. dofetilide 43-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 193-197 11698075-4 2001 These data indicate that a [3H]dofetilide binding assay using HERG membranes may help identify compounds that prolong the QT interval. [3h]dofetilide 27-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 11561083-0 2001 Effects of cocaine and its major metabolites on the HERG-encoded potassium channel. Cocaine 11-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 11561083-3 2001 Cocaine blocked HERG-encoded potassium channels with an IC50 of 4.4 +/- 1.1 microM (22 degrees C). Cocaine 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 11561083-4 2001 Cocaethylene (a metabolite formed in the presence of ethanol) had a significantly lower IC50 of 1.2 +/- 1.1 microM (P < 0.0001), and cocaine"s primary pyrolysis metabolite methylecgonidine blocked HERG with a higher IC50 of 171.7 +/- 1.2 microM. cocaethylene 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-204 11561083-4 2001 Cocaethylene (a metabolite formed in the presence of ethanol) had a significantly lower IC50 of 1.2 +/- 1.1 microM (P < 0.0001), and cocaine"s primary pyrolysis metabolite methylecgonidine blocked HERG with a higher IC50 of 171.7 +/- 1.2 microM. Ethanol 53-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-204 11511086-1 2001 The antipsychotic drugs sertindole and pimozide are known to prolong the QT interval on the electrocardiogram via a high affinity block of the cardiac K(+) channel known as HERG (human ether-a-go-go-related gene; erg1). sertindole 24-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 11511086-1 2001 The antipsychotic drugs sertindole and pimozide are known to prolong the QT interval on the electrocardiogram via a high affinity block of the cardiac K(+) channel known as HERG (human ether-a-go-go-related gene; erg1). sertindole 24-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 213-217 11511086-1 2001 The antipsychotic drugs sertindole and pimozide are known to prolong the QT interval on the electrocardiogram via a high affinity block of the cardiac K(+) channel known as HERG (human ether-a-go-go-related gene; erg1). Pimozide 39-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 11511086-1 2001 The antipsychotic drugs sertindole and pimozide are known to prolong the QT interval on the electrocardiogram via a high affinity block of the cardiac K(+) channel known as HERG (human ether-a-go-go-related gene; erg1). Pimozide 39-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 213-217 11669488-4 2001 Mercury (10 microM) inhibited the K+-channels ROMK and HERG, the phosphate transporter NaPi-3, the amino acid transporter rBAT, the cation transporter OCT-2, and the osmolyte transporter BGT. Mercury 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 11455010-0 2001 Open channel block of HERG K(+) channels by vesnarinone. vesnarinone 44-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 11455010-2 2001 To elucidate the mechanisms, we studied the effects of vesnarinone on HERG, the cloned human I(Kr) channel, heterologously expressed in Xenopus laevis oocytes. vesnarinone 55-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 11455010-3 2001 Vesnarinone caused a concentration-dependent inhibition of HERG currents with an IC(50) value of 17.7 +/- 2.5 microM at 0 mV (n = 6). vesnarinone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-63 11455010-8 2001 Vesnarinone produced similar effects on inactivation-removed mutant (G628C/S631C) HERG channels. vesnarinone 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 11455010-10 2001 Amino acids important for the binding of vesnarinone were identified using alanine-scanning mutagenesis of residues believed to line the inner cavity of the HERG channel. vesnarinone 41-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 157-161 11455010-10 2001 Amino acids important for the binding of vesnarinone were identified using alanine-scanning mutagenesis of residues believed to line the inner cavity of the HERG channel. Alanine 75-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 157-161 11455010-13 2001 In conclusion, vesnarinone preferentially blocks open HERG channels, with little effect on channels in the rested or inactivated state. vesnarinone 15-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 11377395-0 2001 Testosterone-mediated modulation of HERG blockade by proarrhythmic agents. Testosterone 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 11377395-2 2001 Men are known to be at a lower risk for drug-induced TdP than women suggesting a role of sex steroid hormones, androgens and estrogens, in modulation of drug sensitivity of cardiac K(+) channels, particularly those encoded by HERG. Steroids 93-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 226-230 11306689-0 2001 Cocaine blocks HERG, but not KvLQT1+minK, potassium channels. Cocaine 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-19 11306689-2 2001 We investigated the effect of cocaine on the human K(+) channels HERG and KvLQT1+minK that encode native rapidly (I(Kr)) and slowly (I(Ks)) activating delayed rectifier K(+) channels in the heart. Cocaine 30-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 11306689-10 2001 Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. n-methyl-cocaine 6-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 11306689-10 2001 Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. n-methyl-cocaine 6-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 242-246 11306689-10 2001 Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. Cocaine 15-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 11306689-10 2001 Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. Cocaine 15-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 242-246 11306689-10 2001 Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. Cocaine 68-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 11306689-10 2001 Using N-methyl-cocaine, a permanently charged, membrane-impermeable cocaine analog, block of HERG channels rapidly developed when the drug was applied intracellularly through the patch pipette, suggesting that the cocaine binding site on the HERG protein is located on a cytoplasmic accessible domain. Cocaine 68-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 242-246 11306689-11 2001 These results indicate that cocaine suppresses HERG, but not KvLQT1+minK, channels by preferentially blocking activated channels, that it unblocks upon repolarization, and does so with unique ultrarapid kinetics. Cocaine 28-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 11306689-12 2001 Because the cocaine concentration range we studied is achieved in humans, HERG block may provide an additional mechanism for cocaine-induced arrhythmias and sudden death. Cocaine 12-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 11306689-12 2001 Because the cocaine concentration range we studied is achieved in humans, HERG block may provide an additional mechanism for cocaine-induced arrhythmias and sudden death. Cocaine 125-132 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 11693770-8 2001 The epinephrine-induced increases in the mean QTc-e and Tcp-e were larger in LQT1 than in LQT2, and were more pronounced when the averaged data were obtained from 24-leads than from 87-leads. Epinephrine 4-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 11693770-10 2001 The increase in the QTc-eD was larger in LQT1 than in LQT2 patients. qtc-ed 20-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 54-58 11166283-0 2001 [3H]dofetilide binding in SHSY5Y and HEK293 cells expressing a HERG-like K+ channel? Tritium 1-3 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 11166283-0 2001 [3H]dofetilide binding in SHSY5Y and HEK293 cells expressing a HERG-like K+ channel? dofetilide 4-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 11166283-4 2001 Electrophysiological studies showed that SHSY5Y cells contained a HERG-like K+ current blocked by application of dofetilide to either side of the membrane. dofetilide 113-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 11166283-7 2001 That [3H]dofetilide is specific for I(Kr)/HERG may be questionable, as HEK293 and CHO-K1 cells contain no such functional K+ current. [3h]dofetilide 5-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 11741516-7 2001 Berberine blocked the HERG channels potently with an IC50 value of approximately 75 micromol/L. Berberine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 11741516-9 2001 CONCLUSION: Berberine prolonged APD and possessed blocking effect on IK1, IK, and HERG channel expressed in Xenopus oocytes. Berberine 12-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 11741516-10 2001 The antiarrhythmic mechanism of berberine is related to its inhibitory effects on IK1, IK, and HERG channel. Berberine 32-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 95-99 11164846-0 2001 Antiarrhythmic drug carvedilol inhibits HERG potassium channels. Carvedilol 20-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 11164846-6 2001 RESULTS: Carvedilol at a concentration of 10 microM blocked HERG potassium tail currents by 47%. Carvedilol 9-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 11164846-6 2001 RESULTS: Carvedilol at a concentration of 10 microM blocked HERG potassium tail currents by 47%. Potassium 65-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 11164846-9 2001 CONCLUSION: This is the first study demonstrating that carvedilol blocks HERG potassium channels. Carvedilol 55-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 11160646-10 2001 These results suggest that 1) bupivacaine and IQB-9302 block the open state of hKv1.5, Kv2.1, Kv4.3, and HERG channels; and 2) small differences at the N-substituent of these drugs do not affect the drug-induced block of Kv2.1, Kv4.3, or HERG, but specifically modify block of hKv1.5 channels. IQB 9302 46-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 11160646-10 2001 These results suggest that 1) bupivacaine and IQB-9302 block the open state of hKv1.5, Kv2.1, Kv4.3, and HERG channels; and 2) small differences at the N-substituent of these drugs do not affect the drug-induced block of Kv2.1, Kv4.3, or HERG, but specifically modify block of hKv1.5 channels. IQB 9302 46-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 238-242 11160863-3 2001 In this study, cocaine was found to be an inhibitor of HERG channels that underlie the rapidly activating component of I(K). Cocaine 15-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 11046096-7 2000 IC(50) values for block of the HERG cardiac K(+) channel measured 3.73, 0.81, 5.95, and 12.1 microM for granisetron, ondansetron, dolasetron, and MDL 74,156, respectively. Ondansetron 117-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 11046096-7 2000 IC(50) values for block of the HERG cardiac K(+) channel measured 3.73, 0.81, 5.95, and 12.1 microM for granisetron, ondansetron, dolasetron, and MDL 74,156, respectively. dolasetron 130-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 11046096-8 2000 Ondansetron (3 microM) also slowed decay of HERG tail currents. Ondansetron 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 11046096-12 2000 The submicromolar affinity of ondansetron for the HERG K(+) channel likely underlies the prolongation of cardiac repolarization reported for this drug. Ondansetron 30-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 11040340-0 2000 Effects of fluoroquinolones on HERG currents. Fluoroquinolones 11-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 11040340-1 2000 We have investigated the effects of four fluoroquinolones on the human ether-a-go-go-related gene (HERG) mediated K(+) currents to evaluate their potential to induce QT-prolongation. Fluoroquinolones 41-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 11040340-3 2000 Bath application of sparfloxacin, moxifloxacin and grepafloxacin produced an inhibition of HERG outward currents at -40 mV with EC(50) of 13.5+/-0.8, 41. sparfloxacin 20-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11040340-3 2000 Bath application of sparfloxacin, moxifloxacin and grepafloxacin produced an inhibition of HERG outward currents at -40 mV with EC(50) of 13.5+/-0.8, 41. Moxifloxacin 34-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11040340-3 2000 Bath application of sparfloxacin, moxifloxacin and grepafloxacin produced an inhibition of HERG outward currents at -40 mV with EC(50) of 13.5+/-0.8, 41. grepafloxacin 51-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 11009462-9 2000 However, trafficking could not be restored by chemical chaperones or E-4031, a specific blocker of HERG channels. E 4031 69-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 10866950-2 2000 Selective deletion of the HERG-specific sequence (HERG Delta138-373) located between the conserved initial amino terminus (the eag or PAS domain) and the first transmembrane helix accelerates channel activation and shifts its voltage dependence to hyperpolarized values. Methyl 2-acetamido-2-deoxy-beta-D-glucopyranoside 127-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 10866950-2 2000 Selective deletion of the HERG-specific sequence (HERG Delta138-373) located between the conserved initial amino terminus (the eag or PAS domain) and the first transmembrane helix accelerates channel activation and shifts its voltage dependence to hyperpolarized values. Methyl 2-acetamido-2-deoxy-beta-D-glucopyranoside 127-130 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 10866950-2 2000 Selective deletion of the HERG-specific sequence (HERG Delta138-373) located between the conserved initial amino terminus (the eag or PAS domain) and the first transmembrane helix accelerates channel activation and shifts its voltage dependence to hyperpolarized values. Protactinium 134-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 10866950-2 2000 Selective deletion of the HERG-specific sequence (HERG Delta138-373) located between the conserved initial amino terminus (the eag or PAS domain) and the first transmembrane helix accelerates channel activation and shifts its voltage dependence to hyperpolarized values. Protactinium 134-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 10871331-3 2000 HERG channel current was inhibited by vesnarinone in a concentration-dependent manner, whereas KvLQT1/minK current was hardly affected by the drug. vesnarinone 38-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 10871331-4 2000 The inhibition of HERG current by vesnarinone became more prominent and faster as the membrane potential was more depolarized. vesnarinone 34-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 10871331-8 2000 Therefore, the effect of vesnarinone on the HERG-K(+) current could be adequately described by a simple kinetic model of drug-channel interaction. vesnarinone 25-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 10898527-0 2000 Altered gating of HERG potassium channels by cobalt and lanthanum. Cobalt 45-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 10898527-0 2000 Altered gating of HERG potassium channels by cobalt and lanthanum. Lanthanum 56-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 10898527-4 2000 Previous studies have reported that IKr of myocytes, and HERG channels heterologously expressed in Xenopus oocytes, are reduced by external Co2+ and La3+. Cobalt(2+) 140-144 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 10898527-4 2000 Previous studies have reported that IKr of myocytes, and HERG channels heterologously expressed in Xenopus oocytes, are reduced by external Co2+ and La3+. lanthanum(3+) 149-153 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 10898527-5 2000 We have reinvestigated the "blocking" effect of Co2+ and La3+ on HERG channels expressed in Xenopus oocytes. Cobalt(2+) 48-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 10898527-5 2000 We have reinvestigated the "blocking" effect of Co2+ and La3+ on HERG channels expressed in Xenopus oocytes. lanthanum(3+) 57-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 10898527-9 2000 Co2+ and La3+ accelerated the rate of deactivation, decreased the rate of current activation, and shifted the half-point of the HERG channel activation curve by +53 and +65 mV, respectively. Cobalt(2+) 0-4 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 10898527-9 2000 Co2+ and La3+ accelerated the rate of deactivation, decreased the rate of current activation, and shifted the half-point of the HERG channel activation curve by +53 and +65 mV, respectively. lanthanum(3+) 9-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 10828461-1 2000 The S631C mutation in human ether-a-go-go-related gene (HERG) channels has previously been reported to disrupt C-type inactivation and ion-selectivity when Cys-631 is in the oxidized state. Cysteine 156-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 10828461-3 2000 We demonstrate that HERGS631C in its reduced state is fully blocked by 1 microM astemizole, terfenadine and dofetilide, similar to wild-type HERG channels. Astemizole 80-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 10828461-3 2000 We demonstrate that HERGS631C in its reduced state is fully blocked by 1 microM astemizole, terfenadine and dofetilide, similar to wild-type HERG channels. Terfenadine 92-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 10828461-3 2000 We demonstrate that HERGS631C in its reduced state is fully blocked by 1 microM astemizole, terfenadine and dofetilide, similar to wild-type HERG channels. dofetilide 108-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 11723241-2 2001 HERG/I(Kr) channels are blocked selectively by class III antiarrhythmic methanesulfonanilide drugs such as dofetilide. N-Phenylmethanesulfonamide 72-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11602243-1 2001 We did the experiments to search for amino acids that affect quinidine binding to the HERG channel, and have identified an amino acid whose change by mutation affects the binding of various drugs. Quinidine 61-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 11561083-6 2001 Blockade of HERG by cocaine, cocaethylene, and methylecgonidine increased significantly over the voltage range where HERG activates, but became constant at voltages where HERG activation was maximal, indicating that all three drugs block open channels, but by a mechanism that is not highly sensitive to voltage per se. Cocaine 20-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 11561083-6 2001 Blockade of HERG by cocaine, cocaethylene, and methylecgonidine increased significantly over the voltage range where HERG activates, but became constant at voltages where HERG activation was maximal, indicating that all three drugs block open channels, but by a mechanism that is not highly sensitive to voltage per se. cocaethylene 29-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 11561083-6 2001 Blockade of HERG by cocaine, cocaethylene, and methylecgonidine increased significantly over the voltage range where HERG activates, but became constant at voltages where HERG activation was maximal, indicating that all three drugs block open channels, but by a mechanism that is not highly sensitive to voltage per se. anhydroecgonine methyl ester 47-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 11561083-6 2001 Blockade of HERG by cocaine, cocaethylene, and methylecgonidine increased significantly over the voltage range where HERG activates, but became constant at voltages where HERG activation was maximal, indicating that all three drugs block open channels, but by a mechanism that is not highly sensitive to voltage per se. anhydroecgonine methyl ester 47-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 11561083-6 2001 Blockade of HERG by cocaine, cocaethylene, and methylecgonidine increased significantly over the voltage range where HERG activates, but became constant at voltages where HERG activation was maximal, indicating that all three drugs block open channels, but by a mechanism that is not highly sensitive to voltage per se. anhydroecgonine methyl ester 47-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 11561083-8 2001 We conclude that cocaethylene is slightly more potent than cocaine as a blocker of HERG, whereas methylecgonidine has much lower potency, and both benzoylecgonine and ecgonine methyl ester are essentially inactive at clinically relevant concentrations. cocaethylene 17-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 11561083-8 2001 We conclude that cocaethylene is slightly more potent than cocaine as a blocker of HERG, whereas methylecgonidine has much lower potency, and both benzoylecgonine and ecgonine methyl ester are essentially inactive at clinically relevant concentrations. Cocaine 59-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-87 11561091-0 2001 Interactions of the antimalarial drug mefloquine with the human cardiac potassium channels KvLQT1/minK and HERG. Mefloquine 38-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-111 11561091-3 2001 Mefloquine can prolong cardiac repolarization, especially when coadministered with halofantrine, an antagonist of the human ether-a-go-go-related gene (HERG) cardiac K+ channel. Mefloquine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 152-156 11561091-3 2001 Mefloquine can prolong cardiac repolarization, especially when coadministered with halofantrine, an antagonist of the human ether-a-go-go-related gene (HERG) cardiac K+ channel. halofantrine 83-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 152-156 11561091-8 2001 HERG channel currents were about 6-fold less sensitive to block by mefloquine (IC50 = 5.6 microM). Mefloquine 67-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11561091-12 2001 Inhibition by mefloquine of KvLQT1/minK in the human heart may in part explain the synergistic prolongation of QT interval observed when this drug is coadministered with the HERG antagonist halofantrine. Mefloquine 14-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 11561091-12 2001 Inhibition by mefloquine of KvLQT1/minK in the human heart may in part explain the synergistic prolongation of QT interval observed when this drug is coadministered with the HERG antagonist halofantrine. halofantrine 190-202 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-178 11468227-5 2001 Interestingly, six mutations were found in the region of the gene coding for the Per-Arnt-Sim (PAS) and PAS-S1 regions of the HERG protein, stressing the need to examine the entire gene when screening for mutations. Aminosalicylic Acid 95-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 126-130 11377395-3 2001 Here by using neuroleptic agents haloperidol, pimozide, and fluspirilene, all of which can induce TdP, and a steroid hormone-sensitive system Xenopus oocytes for HERG channels expression we show that testosterone is able to reduce HERG-blocking potency of neuroleptics. Pimozide 46-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 11377395-3 2001 Here by using neuroleptic agents haloperidol, pimozide, and fluspirilene, all of which can induce TdP, and a steroid hormone-sensitive system Xenopus oocytes for HERG channels expression we show that testosterone is able to reduce HERG-blocking potency of neuroleptics. Fluspirilene 60-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 11377395-3 2001 Here by using neuroleptic agents haloperidol, pimozide, and fluspirilene, all of which can induce TdP, and a steroid hormone-sensitive system Xenopus oocytes for HERG channels expression we show that testosterone is able to reduce HERG-blocking potency of neuroleptics. Steroids 109-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 11377395-3 2001 Here by using neuroleptic agents haloperidol, pimozide, and fluspirilene, all of which can induce TdP, and a steroid hormone-sensitive system Xenopus oocytes for HERG channels expression we show that testosterone is able to reduce HERG-blocking potency of neuroleptics. Steroids 109-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 231-235 11377395-3 2001 Here by using neuroleptic agents haloperidol, pimozide, and fluspirilene, all of which can induce TdP, and a steroid hormone-sensitive system Xenopus oocytes for HERG channels expression we show that testosterone is able to reduce HERG-blocking potency of neuroleptics. Testosterone 200-212 potassium voltage-gated channel subfamily H member 2 Homo sapiens 162-166 11377395-3 2001 Here by using neuroleptic agents haloperidol, pimozide, and fluspirilene, all of which can induce TdP, and a steroid hormone-sensitive system Xenopus oocytes for HERG channels expression we show that testosterone is able to reduce HERG-blocking potency of neuroleptics. Testosterone 200-212 potassium voltage-gated channel subfamily H member 2 Homo sapiens 231-235 11377395-4 2001 Haloperidol, pimozide, and fluspirilene inhibited HERG current with IC(50) of 1.36, 1.74, and 2.34 microM, and maximal block of 73%, 76% and 65%, respectively. Haloperidol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 11377395-4 2001 Haloperidol, pimozide, and fluspirilene inhibited HERG current with IC(50) of 1.36, 1.74, and 2.34 microM, and maximal block of 73%, 76% and 65%, respectively. Pimozide 13-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 11377395-4 2001 Haloperidol, pimozide, and fluspirilene inhibited HERG current with IC(50) of 1.36, 1.74, and 2.34 microM, and maximal block of 73%, 76% and 65%, respectively. Fluspirilene 27-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 11377395-6 2001 Pretreatment of HERG-expressing oocytes with 1 microM testosterone increased the IC(50) values to 2.73, 2.08, and 5.04 microM, reduced the maximal block to 65%, 59%, and 64%, and strongly diminished voltage-dependence of the blockade. Testosterone 54-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 11377395-7 2001 Testosterone treatment per se produced about a 35% reduction of HERG current compared with untreated oocytes. Testosterone 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 11404399-3 2001 Human ether-a-go-go-related gene (HERG) K(+) channels are unusually sensitive to external calcium concentration ([Ca(2+)](o)). Calcium 90-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 11404399-5 2001 The HERG-calcium concentration-response relationship spans the physiological range for [Ca(2+)](o). Calcium 9-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 11278781-0 2001 Analysis of the cyclic nucleotide binding domain of the HERG potassium channel and interactions with KCNE2. Nucleotides, Cyclic 16-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 11278781-1 2001 Mutations in the cyclic nucleotide binding domain (CNBD) of the human ether-a-go-go-related gene (HERG) K+ channel are associated with LQT2, a form of hereditary Long QT syndrome (LQTS). Nucleotides, Cyclic 17-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 11278781-1 2001 Mutations in the cyclic nucleotide binding domain (CNBD) of the human ether-a-go-go-related gene (HERG) K+ channel are associated with LQT2, a form of hereditary Long QT syndrome (LQTS). Nucleotides, Cyclic 17-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 135-139 11278781-2 2001 Elevation of cAMP can modulate HERG K+ channels both by direct binding and indirect regulation through protein kinase A. Cyclic AMP 13-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 11278781-3 2001 To assess the physiological significance of cAMP binding to HERG, we introduced mutations to disrupt the cyclic nucleotide binding domain. Cyclic AMP 44-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 11259532-0 2001 Hydrogen peroxide modifies the kinetics of HERG channel expressed in a mammalian cell line. Hydrogen Peroxide 0-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 11259532-3 2001 As these effects could involve the modulation of repolarizing currents, we assessed effects of H2O2 on HERG (which encodes the cardiac potassium channel I(Kr)) expressed in Chinese hamster ovary cells. Hydrogen Peroxide 95-99 potassium voltage-gated channel subfamily H member 2 Homo sapiens 103-107 11259532-5 2001 HERG activation and deactivation were accelerated when cells were superfused with 30 microM, 100 microM, or 1 mM H2O2. Hydrogen Peroxide 113-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11259532-9 2001 This indicates that H2O2 diffuses intracellularly before acting on HERG and that its effects on activation but not deactivation are mediated by the superoxide anion. Hydrogen Peroxide 20-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 11238279-7 2001 HERG current in oocytes was reduced by amiodarone (IC(50)=38 micromol/L), whereas KvLQT1/minK current was unaffected by 300 micromol/L amiodarone. Amiodarone 39-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11741516-0 2001 Inhibitory effects of berberine on IK1, IK, and HERG channels of cardiac myocytes. Berberine 22-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 11160646-9 2001 Bupivacaine and IQB-9302 induced a similar degree of block of HERG channels and induced a steep voltage-dependent decrease of the relative current. Bupivacaine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 11160646-9 2001 Bupivacaine and IQB-9302 induced a similar degree of block of HERG channels and induced a steep voltage-dependent decrease of the relative current. IQB 9302 16-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 11160646-10 2001 These results suggest that 1) bupivacaine and IQB-9302 block the open state of hKv1.5, Kv2.1, Kv4.3, and HERG channels; and 2) small differences at the N-substituent of these drugs do not affect the drug-induced block of Kv2.1, Kv4.3, or HERG, but specifically modify block of hKv1.5 channels. Bupivacaine 30-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 11160646-10 2001 These results suggest that 1) bupivacaine and IQB-9302 block the open state of hKv1.5, Kv2.1, Kv4.3, and HERG channels; and 2) small differences at the N-substituent of these drugs do not affect the drug-induced block of Kv2.1, Kv4.3, or HERG, but specifically modify block of hKv1.5 channels. Bupivacaine 30-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 238-242 11125032-0 2001 Interactions of a series of fluoroquinolone antibacterial drugs with the human cardiac K+ channel HERG. Fluoroquinolones 28-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 98-102 11125032-7 2001 Block of HERG by sparfloxacin displayed a positive voltage dependence. sparfloxacin 17-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 11125032-9 2001 These results provide a mechanism for the QT prolongation observed clinically with administration of sparfloxacin and certain other fluoroquinolones because free plasma levels of these drugs after therapeutic doses approximate those concentrations that inhibit HERG channel current. sparfloxacin 101-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 261-265 11125032-9 2001 These results provide a mechanism for the QT prolongation observed clinically with administration of sparfloxacin and certain other fluoroquinolones because free plasma levels of these drugs after therapeutic doses approximate those concentrations that inhibit HERG channel current. Fluoroquinolones 132-148 potassium voltage-gated channel subfamily H member 2 Homo sapiens 261-265 11125032-10 2001 In the cases of levofloxacin, ciprofloxacin, and ofloxacin, inhibition of HERG occurs at concentrations much greater than those observed clinically. Levofloxacin 16-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 11125032-10 2001 In the cases of levofloxacin, ciprofloxacin, and ofloxacin, inhibition of HERG occurs at concentrations much greater than those observed clinically. Ciprofloxacin 30-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 11125032-10 2001 In the cases of levofloxacin, ciprofloxacin, and ofloxacin, inhibition of HERG occurs at concentrations much greater than those observed clinically. Ofloxacin 19-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 11125032-12 2001 HERG channel affinity should be an important criterion for the development of newer fluoroquinolones. Fluoroquinolones 84-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 11575008-9 2001 On the other hand terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (330 and 480 nmol/L, respectively), whereas loratadine was about 300-fold less potent. Terfenadine 18-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 11575008-9 2001 On the other hand terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (330 and 480 nmol/L, respectively), whereas loratadine was about 300-fold less potent. Astemizole 34-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 11090546-0 2000 Probing the interaction between inactivation gating and Dd-sotalol block of HERG. Fumigant 93 56-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 11090546-0 2000 Probing the interaction between inactivation gating and Dd-sotalol block of HERG. Sotalol 59-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 11090546-1 2000 Potassium channels encoded by HERG underlie I:(Kr), a sensitive target for most class III antiarrhythmic drugs, including methanesulfonanilides such as Dd-sotalol. methanesulfonanilides 122-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 11090546-1 2000 Potassium channels encoded by HERG underlie I:(Kr), a sensitive target for most class III antiarrhythmic drugs, including methanesulfonanilides such as Dd-sotalol. dd-sotalol 152-162 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 11090546-5 2000 In the present study, we identify a definitive role for inactivation gating in Dd-sotalol block of HERG, using interventions complementary to mutagenesis. Fumigant 93 79-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 11090546-5 2000 In the present study, we identify a definitive role for inactivation gating in Dd-sotalol block of HERG, using interventions complementary to mutagenesis. Sotalol 82-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 11090546-7 2000 In normal extracellular solutions, block of HERG current by 300 micromol/L Dd-sotalol reached 80% after a 10-minute period of repetitive depolarization to +20 mV. dd-sotalol 75-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 11090546-8 2000 Maneuvers that impeded steady-state inactivation also reduced Dd-sotalol block of HERG: 100 micromol/L Cd(2+) reduced steady-state block to 55% at +20 mV (P:<0.05); removing extracellular Na(+) reduced block to 44% (P:<0.05). dd-sotalol 62-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 11082114-0 2000 Inhibition of HERG1 K(+) channels by the novel second-generation antihistamine mizolastine. antihistamine mizolastine 65-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-19 11082114-5 2000 In the present study, the potential blocking ability of the novel second-generation H(1) receptor antagonist mizolastine of the HERG1 K(+) channels heterologously expressed in Xenopus oocytes and in HEK 293 cells or constitutively present in SH-SY5Y human neuroblastoma cells has been examined and compared to that of astemizole. mizolastine 109-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-133 11082114-5 2000 In the present study, the potential blocking ability of the novel second-generation H(1) receptor antagonist mizolastine of the HERG1 K(+) channels heterologously expressed in Xenopus oocytes and in HEK 293 cells or constitutively present in SH-SY5Y human neuroblastoma cells has been examined and compared to that of astemizole. Astemizole 318-328 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-133 11082114-7 2000 Mizolastine blocked HERG1 K(+) channels expressed in Xenopus oocytes with an estimated IC(50) of 3.4 microM. mizolastine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-25 11082114-10 2000 In human embryonic kidney 293 cells (HEK 293 cells) stably transfected with HERG1 cDNA, extracellular application of mizolastine exerted a dose-related inhibitory action on I(HERG1), with an IC(50) of 350+/-76 nM. mizolastine 117-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-81 11082114-10 2000 In human embryonic kidney 293 cells (HEK 293 cells) stably transfected with HERG1 cDNA, extracellular application of mizolastine exerted a dose-related inhibitory action on I(HERG1), with an IC(50) of 350+/-76 nM. mizolastine 117-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 175-180 11082114-11 2000 Furthermore, mizolastine dose-dependently inhibited HERG1 K(+) channels constitutively expressed in SH-SY5Y human neuroblastoma clonal cells. mizolastine 13-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-57 11082114-13 2000 The results of the present study suggest that the novel second-generation H(1) receptor antagonist mizolastine, in concentrations higher than those achieved in vivo during standard therapy, is able to block in some degree both constitutively and heterologously expressed HERG1 K(+) channels, and confirm the heterogeneity of molecules belonging to this therapeutical class with respect to their HERG1-inhibitory action. mizolastine 99-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 271-276 11082114-13 2000 The results of the present study suggest that the novel second-generation H(1) receptor antagonist mizolastine, in concentrations higher than those achieved in vivo during standard therapy, is able to block in some degree both constitutively and heterologously expressed HERG1 K(+) channels, and confirm the heterogeneity of molecules belonging to this therapeutical class with respect to their HERG1-inhibitory action. mizolastine 99-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 395-400 11033107-0 2000 Comparative effects of azimilide and ambasilide on the human ether-a-go-go-related gene (HERG) potassium channel. azimilide 23-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 11033107-0 2000 Comparative effects of azimilide and ambasilide on the human ether-a-go-go-related gene (HERG) potassium channel. ambasilide 37-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 11033107-1 2000 OBJECTIVE: To evaluate the effects of azimilide and ambasilide on the biophysical properties of the human-ether-a-go-go-related (HERG) channel. azimilide 38-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-127 11033107-1 2000 OBJECTIVE: To evaluate the effects of azimilide and ambasilide on the biophysical properties of the human-ether-a-go-go-related (HERG) channel. azimilide 38-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 129-133 11033107-1 2000 OBJECTIVE: To evaluate the effects of azimilide and ambasilide on the biophysical properties of the human-ether-a-go-go-related (HERG) channel. ambasilide 52-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-127 11033107-1 2000 OBJECTIVE: To evaluate the effects of azimilide and ambasilide on the biophysical properties of the human-ether-a-go-go-related (HERG) channel. ambasilide 52-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 129-133 11033107-7 2000 By comparison, ambasilide inhibited HERG channels with lower potency (IC(50) 3.6 microM), in a voltage- and time-dependent but frequency-independent manner (0.03-1 Hz). ambasilide 15-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 36-40 11033107-10 2000 CONCLUSIONS: Inhibition of HERG channels by azimilide and ambasilide exhibits a similar time and voltage-dependence. azimilide 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 11033107-10 2000 CONCLUSIONS: Inhibition of HERG channels by azimilide and ambasilide exhibits a similar time and voltage-dependence. ambasilide 58-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 10931813-4 2000 Isoproterenol (50 to 100 nmol/L) was used to mimic an increase in beta-adrenergic tone, d-sotalol (100 micromol/L) to block I(Kr) (LQT2 model), and ATX-II (20 nmol/L) to augment late I(Na) (LQT3 model). Isoproterenol 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10931813-7 2000 High concentrations (10 to 20 micromol/L) completely reversed the effects of 293B+/-isoproterenol and those of d-sotalol to increase APD(90) and TDR and to induce TdP in LQT1 and LQT2 models. Isoproterenol 84-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 10931813-7 2000 High concentrations (10 to 20 micromol/L) completely reversed the effects of 293B+/-isoproterenol and those of d-sotalol to increase APD(90) and TDR and to induce TdP in LQT1 and LQT2 models. Dexsotalol 111-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 10952689-0 2000 Inhibition of HERG potassium channels by the antimalarial agent halofantrine. halofantrine 64-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 10952689-4 2000 We examined the effects of halofantrine on HERG potassium channels stably expressed in Chinese hamster ovary (CHO-K1) cells. halofantrine 27-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 10952689-5 2000 Halofantrine blocked HERG tail currents elicited on repolarization to -60 mV from +30 mV with an IC(50) of 196.9 nM. halofantrine 0-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 10952689-10 2000 We conclude that HERG channel inhibition by halofantrine is the likely underlying cellular mechanism for QT prolongation. halofantrine 44-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 10896755-0 2000 State-dependent barium block of wild-type and inactivation-deficient HERG channels in Xenopus oocytes. Barium 16-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 10896755-1 2000 The effects of Ba2+ on current resulting from the heterologous expression of the human ether-a-go-go related gene (HERG) (IHERG) was studied with two-electrode voltage clamp techniques in Xenopus oocytes. N-methyl-valyl-amiclenomycin 15-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 11714889-4 2001 We compared the effects of four different 5-HT4 receptor agonists (cisapride, prucalopride, renzapride and mosapride) on cloned HERG channels with the objective to evaluate and compare their proarrhythmic potential. Cisapride 67-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 11714889-4 2001 We compared the effects of four different 5-HT4 receptor agonists (cisapride, prucalopride, renzapride and mosapride) on cloned HERG channels with the objective to evaluate and compare their proarrhythmic potential. prucalopride 78-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 11714889-4 2001 We compared the effects of four different 5-HT4 receptor agonists (cisapride, prucalopride, renzapride and mosapride) on cloned HERG channels with the objective to evaluate and compare their proarrhythmic potential. renzapride 92-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 128-132 11714889-7 2001 Cisapride inhibited the HERG channels in a concentration-dependent manner with an IC(50) of 2.4 10(-7) M. The IC(50) value for prucalopride to block HERG (5.7 10(-6) M) was 20-fold higher than that of cisapride. Cisapride 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 11714889-7 2001 Cisapride inhibited the HERG channels in a concentration-dependent manner with an IC(50) of 2.4 10(-7) M. The IC(50) value for prucalopride to block HERG (5.7 10(-6) M) was 20-fold higher than that of cisapride. Cisapride 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 11714889-7 2001 Cisapride inhibited the HERG channels in a concentration-dependent manner with an IC(50) of 2.4 10(-7) M. The IC(50) value for prucalopride to block HERG (5.7 10(-6) M) was 20-fold higher than that of cisapride. prucalopride 127-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 11714889-7 2001 Cisapride inhibited the HERG channels in a concentration-dependent manner with an IC(50) of 2.4 10(-7) M. The IC(50) value for prucalopride to block HERG (5.7 10(-6) M) was 20-fold higher than that of cisapride. prucalopride 127-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 149-153 11714889-7 2001 Cisapride inhibited the HERG channels in a concentration-dependent manner with an IC(50) of 2.4 10(-7) M. The IC(50) value for prucalopride to block HERG (5.7 10(-6) M) was 20-fold higher than that of cisapride. Cisapride 201-210 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 11714889-12 2001 We conclude that the rank order of potency of 5-HT4 agonists to block HERG is cisapride > renzapride > prucalopride > mosapride. Cisapride 78-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 11714889-12 2001 We conclude that the rank order of potency of 5-HT4 agonists to block HERG is cisapride > renzapride > prucalopride > mosapride. renzapride 93-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 11714889-12 2001 We conclude that the rank order of potency of 5-HT4 agonists to block HERG is cisapride > renzapride > prucalopride > mosapride. prucalopride 109-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 11714889-12 2001 We conclude that the rank order of potency of 5-HT4 agonists to block HERG is cisapride > renzapride > prucalopride > mosapride. mosapride 127-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 70-74 10720411-1 2000 The N629D mutation, adjacent to the GFG signature sequence of the HERG1 A K(+) channel, causes long-QT syndrome (LQTS). guanfu base G 36-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-71 10742304-0 2000 Blockade of the HERG human cardiac K(+) channel by the antidepressant drug amitriptyline. Amitriptyline 75-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 10742304-3 2000 We studied the effects of amitriptyline on the human ether-a-go-go-related gene (HERG) channel expressed in Xenopus oocytes and on the rapidly activating delayed rectifier K(+) current (I(Kr)) in rat atrial myocytes. Amitriptyline 26-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 10742304-5 2000 The amplitudes of steady-state currents and tail currents of HERG were decreased by amitriptyline dose-dependently. Amitriptyline 84-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-65 10742304-6 2000 The decrease became more pronounced at more positive potential, suggesting that the block of HERG by amitriptyline is voltage dependent. Amitriptyline 101-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 10742304-7 2000 IC(50) for amitriptyline block of HERG current was progressively decreased according to depolarization: IC(50) values at -30, -10, +10 and +30 mV were 23.0, 8.71, 5.96 and 4.66 microM, respectively. Amitriptyline 11-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 10742304-9 2000 Block of HERG by amitriptyline was use dependent: exhibiting a much faster block at higher activation frequency. Amitriptyline 17-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 10742304-14 2000 78 microM in 4 mM [K(+)](o), suggesting that the affinity of amitriptyline on HERG was decreased by external K(+). Amitriptyline 61-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 10742304-19 2000 In summary, the data suggest that the block of HERG currents may contribute to arrhythmogenic side effects of amitriptyline. Amitriptyline 110-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 10762666-0 2000 High affinity blockade of the HERG cardiac K(+) channel by the neuroleptic pimozide. Pimozide 75-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 10762666-3 2000 To elucidate the mechanism behind these clinical findings, we examined the effects of pimozide on the cloned human cardiac K(+) channels HERG (human ether-a-go-go-related gene; rapid component of delayed rectifier), Kv1.5 (ultra-rapid delayed rectifier) and KvLQT1/minK (slow component of delayed rectifier). Pimozide 86-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 10762666-4 2000 Using patch clamp electrophysiology, we found that pimozide was a potent inhibitor of HERG displaying an IC(50) value of 18 nM. Pimozide 51-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 10762666-6 2000 We conclude that pimozide is a specific, high affinity antagonist of HERG, and that this interaction leads to prolongation of cardiac repolarization. Pimozide 17-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 10716483-8 2000 In LQT2 isoproterenol initially prolonged, then abbreviated, the APD90 of M but always abbreviated EPI, thus transiently increasing TDR and the incidence of TdP. Isoproterenol 8-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 10735633-2 2000 The syndrome was associated with a novel KCNH2 missense mutation, G572R, causing the substitution of a glycine residue at position 572, at the end of the S5 transmembrane segment of the HERG K(+)-channel, with an arginine residue. Glycine 103-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-46 10712445-0 2000 HERG-Like potassium current regulates the resting membrane potential in glomus cells of the rabbit carotid body. Potassium 10-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 10712445-7 2000 The HERG-like current was blocked by dofetilide (DOF) in a concentration-dependent manner (IC(50) = 13 +/- 4 nM, mean +/- SE) and high concentrations of Ba(2+) (1 and 10 mM). dofetilide 37-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10712445-7 2000 The HERG-like current was blocked by dofetilide (DOF) in a concentration-dependent manner (IC(50) = 13 +/- 4 nM, mean +/- SE) and high concentrations of Ba(2+) (1 and 10 mM). dofetilide 49-52 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10712445-7 2000 The HERG-like current was blocked by dofetilide (DOF) in a concentration-dependent manner (IC(50) = 13 +/- 4 nM, mean +/- SE) and high concentrations of Ba(2+) (1 and 10 mM). N-methyl-valyl-amiclenomycin 153-159 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10735633-2 2000 The syndrome was associated with a novel KCNH2 missense mutation, G572R, causing the substitution of a glycine residue at position 572, at the end of the S5 transmembrane segment of the HERG K(+)-channel, with an arginine residue. Glycine 103-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 186-190 10735633-2 2000 The syndrome was associated with a novel KCNH2 missense mutation, G572R, causing the substitution of a glycine residue at position 572, at the end of the S5 transmembrane segment of the HERG K(+)-channel, with an arginine residue. Arginine 213-221 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-46 10735633-2 2000 The syndrome was associated with a novel KCNH2 missense mutation, G572R, causing the substitution of a glycine residue at position 572, at the end of the S5 transmembrane segment of the HERG K(+)-channel, with an arginine residue. Arginine 213-221 potassium voltage-gated channel subfamily H member 2 Homo sapiens 186-190 10860024-3 2000 HERG is blocked by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM) and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Bepridil 19-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 10604956-10 2000 However, both drugs potently blocked HERG current amplitude, with a mean IC(50) of 173 nM for loratadine and 204 nM for terfenadine (pacing rate, 0.1 Hz). Loratadine 94-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 10604956-10 2000 However, both drugs potently blocked HERG current amplitude, with a mean IC(50) of 173 nM for loratadine and 204 nM for terfenadine (pacing rate, 0.1 Hz). Terfenadine 120-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 10860024-3 2000 HERG is blocked by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM) and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Verapamil 50-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 10860024-3 2000 HERG is blocked by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM) and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Mibefradil 85-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 10860024-3 2000 HERG is blocked by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM) and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Diltiazem 143-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 10545354-1 1999 We have studied the functional effects of extracellular Cd(2+) on human ether-a-go-go-related gene (HERG) encoded K(+) channels. Cadmium 56-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 10600781-3 1999 The HERG K(+) channel blockers E-4031 (1 microM), cisapride (1 microM), and La(3+) (100 microM) strongly inhibited these currents as did millimolar concentrations of Ba(2+). E 4031 31-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10600781-3 1999 The HERG K(+) channel blockers E-4031 (1 microM), cisapride (1 microM), and La(3+) (100 microM) strongly inhibited these currents as did millimolar concentrations of Ba(2+). Cisapride 50-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10600781-3 1999 The HERG K(+) channel blockers E-4031 (1 microM), cisapride (1 microM), and La(3+) (100 microM) strongly inhibited these currents as did millimolar concentrations of Ba(2+). lanthanum(3+) 76-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10600781-3 1999 The HERG K(+) channel blockers E-4031 (1 microM), cisapride (1 microM), and La(3+) (100 microM) strongly inhibited these currents as did millimolar concentrations of Ba(2+). Barium 166-168 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 10636190-9 1999 Finally, HERG K+ current elicited in HEK293 cells expressing high levels of HERG protein was decreased 50% by droperidol 32.2 nmol/L. Droperidol 110-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 10636190-9 1999 Finally, HERG K+ current elicited in HEK293 cells expressing high levels of HERG protein was decreased 50% by droperidol 32.2 nmol/L. Droperidol 110-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 10570058-0 1999 Modulation of the K(+) channels encoded by the human ether-a-gogo-related gene-1 (hERG1) by nitric oxide. Nitric Oxide 92-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-87 10570058-1 1999 The inhibition of nitric oxide synthase by N-nitro-L-arginine methyl ester (0.03-3 mM) dose-dependently reduced nitric oxide (NO(*)) levels and enhanced the outward currents carried by human ether-a-gogo-related gene-1 (hERG1) K(+) channels expressed in Xenopus laevis oocytes, whereas the increase in NO(*) levels achieved by exposure to L-arginine (0.03-10 mM) inhibited these currents. Nitric Oxide 18-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 220-225 10570058-1 1999 The inhibition of nitric oxide synthase by N-nitro-L-arginine methyl ester (0.03-3 mM) dose-dependently reduced nitric oxide (NO(*)) levels and enhanced the outward currents carried by human ether-a-gogo-related gene-1 (hERG1) K(+) channels expressed in Xenopus laevis oocytes, whereas the increase in NO(*) levels achieved by exposure to L-arginine (0.03-10 mM) inhibited these currents. n-nitro-l-arginine methyl ester 43-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 220-225 10570058-1 1999 The inhibition of nitric oxide synthase by N-nitro-L-arginine methyl ester (0.03-3 mM) dose-dependently reduced nitric oxide (NO(*)) levels and enhanced the outward currents carried by human ether-a-gogo-related gene-1 (hERG1) K(+) channels expressed in Xenopus laevis oocytes, whereas the increase in NO(*) levels achieved by exposure to L-arginine (0.03-10 mM) inhibited these currents. Arginine 51-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 220-225 10570058-4 1999 The inhibitory effect of NO(*) donors on hERG1 K(+) channels was prevented by the NO(*) scavengers 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide and hemoglobin. 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide 99-153 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-46 10570058-6 1999 Both L-arginine (10 mM) and NOC-18 (0.3 mM) counteracted the stimulatory effect on hERG1 outward currents induced by the radical oxygen species-generating system FeSO(4) (25 microM)/ascorbic acid (50 microM; Fe/Asc). Arginine 5-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-88 10570058-6 1999 Both L-arginine (10 mM) and NOC-18 (0.3 mM) counteracted the stimulatory effect on hERG1 outward currents induced by the radical oxygen species-generating system FeSO(4) (25 microM)/ascorbic acid (50 microM; Fe/Asc). NOC 18 28-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-88 10570058-6 1999 Both L-arginine (10 mM) and NOC-18 (0.3 mM) counteracted the stimulatory effect on hERG1 outward currents induced by the radical oxygen species-generating system FeSO(4) (25 microM)/ascorbic acid (50 microM; Fe/Asc). Oxygen 129-135 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-88 10570058-6 1999 Both L-arginine (10 mM) and NOC-18 (0.3 mM) counteracted the stimulatory effect on hERG1 outward currents induced by the radical oxygen species-generating system FeSO(4) (25 microM)/ascorbic acid (50 microM; Fe/Asc). fesoterodine 162-166 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-88 10570058-6 1999 Both L-arginine (10 mM) and NOC-18 (0.3 mM) counteracted the stimulatory effect on hERG1 outward currents induced by the radical oxygen species-generating system FeSO(4) (25 microM)/ascorbic acid (50 microM; Fe/Asc). Ascorbic Acid 182-195 potassium voltage-gated channel subfamily H member 2 Homo sapiens 83-88 10570058-8 1999 Collectively, the present results suggest that NO(*), both endogenously produced and pharmacologically delivered, may exert in a cGMP-independent way an inhibitory effect on hERG1 outward K(+) currents via an interaction with radical oxygen species either generated under resting conditions or triggered by Fe/Asc. Oxygen 234-240 potassium voltage-gated channel subfamily H member 2 Homo sapiens 174-179 10690305-12 1999 CONCLUSIONS: The data revealed that arginine at position 534 in the S4 region of HERG is indeed involved in voltage-dependence of channel activation as a voltage sensor. Arginine 36-44 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 10388757-1 1999 The proton and Zn2+ effects on the human ether-a-go-go related gene (HERG) channels were studied after expression in Xenopus oocytes and stable transfection in the mammalian L929 cell line. Zinc 15-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 10531299-5 1999 We further show that the antiarrhythmic drug E-4031, which selectively blocks HERG channels, also corrects defective protein trafficking of the N470D mutant and can restore the generation of HERG current. E 4031 45-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 10531299-5 1999 We further show that the antiarrhythmic drug E-4031, which selectively blocks HERG channels, also corrects defective protein trafficking of the N470D mutant and can restore the generation of HERG current. E 4031 45-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 191-195 10531299-7 1999 The effect of E-4031 on HERG protein trafficking was concentration-dependent and required low drug concentrations (saturation present at 5 microM), developed rapidly with drug exposure, and occurred post-translationally. E 4031 14-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 10510456-0 1999 Inhibition of the human ether-a-go-go-related gene (HERG) potassium channel by cisapride: affinity for open and inactivated states. Cisapride 79-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-56 10510456-3 1999 2 In a chronic transfection model using CHO-K1 cells, cisapride inhibited HERG tail currents after a step to +25 mV with similar potency at room and physiological temperatures (IC50 16. Cisapride 54-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 10510456-10 1999 6 In conclusion, HERG channel inhibition by cisapride exhibits features consistent with open and inactivated state binding and is sensitive to external potassium concentration. Cisapride 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 10510456-10 1999 6 In conclusion, HERG channel inhibition by cisapride exhibits features consistent with open and inactivated state binding and is sensitive to external potassium concentration. Potassium 152-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 10413451-1 1999 The modulation of herg gene and HERG currents (I(HERG)) was studied in SH-SY5Y neuroblastoma (NB) cells treated with all-trans-retinoic acid (RA) in the absence or presence of the neurotrophin brain-derived neurotrophic factor (BDNF). Tretinoin 142-144 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 10413451-3 1999 Differentiation of NB cells was accompanied by an increase in herg gene transcription, which attained its maximum after 6 days of treatment with RA and was not further increased by BDNF. Tretinoin 145-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 10413451-4 1999 This effect evidently reflected on HERG currents: In fact, RA produced an increase in HERG current density which was strongly potentiated by BDNF. Tretinoin 59-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 10413451-4 1999 This effect evidently reflected on HERG currents: In fact, RA produced an increase in HERG current density which was strongly potentiated by BDNF. Tretinoin 59-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 10413451-5 1999 Moreover, RA treatment affected the biophysical properties of I(HERG), inducing an increase in the deactivation time constant and a left shift of the activation curve. Tretinoin 10-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 10516162-6 1999 The following were ruled out as the underlying mechanisms: 1) voltage shift in channel activation, 2) pore blockade by protons, 3) protonation of histidines on the extracellular domain of HERG, 4) acceleration of recovery from C-type inactivation, and 5) interaction between an external H(+) binding site and the cytoplasmic NH(2)-terminal domain (a key determinant of HERG deactivation rate). Histidine 146-156 potassium voltage-gated channel subfamily H member 2 Homo sapiens 188-192 10516162-7 1999 Extracellular application of diethylpyrocarbonate caused an irreversible acceleration of HERG deactivation and prevented further acceleration by external acidification. Diethyl Pyrocarbonate 29-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 10488078-4 1999 Application of the cAMP-specific phosphodiesterase (PDE IV) inhibitor Ro-20-1724 (100 microM), which results in an increased cAMP level and PKA stimulation, induced a reduction of HERG wild type outward currents by 19.1% due to a shift in the activation curve of 12.4 mV. Cyclic AMP 19-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 10488078-4 1999 Application of the cAMP-specific phosphodiesterase (PDE IV) inhibitor Ro-20-1724 (100 microM), which results in an increased cAMP level and PKA stimulation, induced a reduction of HERG wild type outward currents by 19.1% due to a shift in the activation curve of 12.4 mV. 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 10488078-4 1999 Application of the cAMP-specific phosphodiesterase (PDE IV) inhibitor Ro-20-1724 (100 microM), which results in an increased cAMP level and PKA stimulation, induced a reduction of HERG wild type outward currents by 19.1% due to a shift in the activation curve of 12.4 mV. Cyclic AMP 125-129 potassium voltage-gated channel subfamily H member 2 Homo sapiens 180-184 10488078-6 1999 Furthermore, the adenylate cyclase activator forskolin that leads to PKA activation (400 microM, 60 min), shifted HERG wild type channel activation by 14.1 mV and reduced currents by 39.9%, whereas HERG 4M channels showed only a small shift of 4.3 mV and a weaker current reduction of 22.3%. Colforsin 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 114-118 10488078-6 1999 Furthermore, the adenylate cyclase activator forskolin that leads to PKA activation (400 microM, 60 min), shifted HERG wild type channel activation by 14.1 mV and reduced currents by 39.9%, whereas HERG 4M channels showed only a small shift of 4.3 mV and a weaker current reduction of 22.3%. Colforsin 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 198-202 10484431-0 1999 Inactivation gating determines nicotine blockade of human HERG channels. Nicotine 31-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-62 10484431-2 1999 To shed some light on the mechanisms of interaction between nicotine and channels, we performed detailed analysis on the human ether-a-go-go-related gene (HERG) channels, which are believed to be equivalent to the native I(Kr) when expressed in Xenopus oocytes. Nicotine 60-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-159 10484431-3 1999 Nicotine suppressed the HERG channels in a concentration-dependent manner with greater potency with voltage protocols, which favor channel inactivation. Nicotine 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-28 10484431-7 1999 Moreover, nicotine lost its ability to block the HERG channels when a single mutation was introduced to a residue located after transmembrane domain 6 (S631A) to remove the rapid channel inactivation. Nicotine 10-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 10484431-8 1999 Our data suggest that the inactivation gating determines nicotine blockade of the HERG channels. Nicotine 57-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 82-86 10560244-6 1999 A C-->T transition in codon 614, leading to substitution of a valine for an alanine residue in the pore region of the HERG protein, was identified. Valine 65-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 10560244-6 1999 A C-->T transition in codon 614, leading to substitution of a valine for an alanine residue in the pore region of the HERG protein, was identified. Alanine 79-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 121-125 10388757-11 1999 We conclude that protons and Zn2+ directly interact with HERG channels and that the interaction results, preferentially, in the regulation of channel deactivation mechanism. Zinc 29-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 10444235-4 1999 In fact, both terfenadine and astemizole have been shown to block HERG K+ channels in a concentration range similar to that found in the plasma of subjects with cardiotoxic manifestations. Terfenadine 14-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 10444235-4 1999 In fact, both terfenadine and astemizole have been shown to block HERG K+ channels in a concentration range similar to that found in the plasma of subjects with cardiotoxic manifestations. Astemizole 30-40 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 10444235-6 1999 In fact, other molecules such as cetirizine, loratadine, acrivastine, and fexofenadine seem to lack both cardiotoxic potential and HERG-blocking ability at therapeutically relevant concentrations. fexofenadine 74-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10096894-1 1999 We have investigated actions of various divalent cations (Ba2+, Sr2+, Mn2+, Co2+, Ni2+, Zn2+) on human ether-a-go-go related gene (HERG) channels expressed in Xenopus laevis oocytes using the voltage clamp technique. N-methyl-valyl-amiclenomycin 58-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10422790-1 1999 Terfenadine and astemizole rarely cause cardiac arrhythmias by suppressing the cardiac rapid delayed rectifier K+ channel encoded by the human ether-a-go-go-related gene (HERG). Terfenadine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 10422790-1 1999 Terfenadine and astemizole rarely cause cardiac arrhythmias by suppressing the cardiac rapid delayed rectifier K+ channel encoded by the human ether-a-go-go-related gene (HERG). Astemizole 16-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 10422790-3 1999 We have therefore compared the effects of epinastine, terfenadine and astemizole on HERG channels expressed in Xenopus oocytes. Astemizole 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 10422790-4 1999 Terfenadine and astemizole suppressed the HERG current with IC50 of 431 nM and 69 nM, respectively. Terfenadine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 10422790-4 1999 Terfenadine and astemizole suppressed the HERG current with IC50 of 431 nM and 69 nM, respectively. Astemizole 16-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 10422790-5 1999 In contrast, 100 microM epinastine inhibited the HERG current by only 11+/-2.1%. epinastine 24-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 10325236-0 1999 Mechanism of block and identification of the verapamil binding domain to HERG potassium channels. Verapamil 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 10325236-2 1999 We studied the effects of verapamil, diltiazem, and nifedipine on HERG K+ channels that encode IKr in native heart cells. Nifedipine 52-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 10325236-3 1999 In our experiments, verapamil caused high-affinity block of HERG current (IC50=143.0 nmol/L), a value close to those reported for verapamil block of L-type Ca2+ channels, whereas diltiazem weakly blocked HERG current (IC50=17.3 micromol/L), and nifedipine did not block HERG current. Verapamil 20-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 10325236-3 1999 In our experiments, verapamil caused high-affinity block of HERG current (IC50=143.0 nmol/L), a value close to those reported for verapamil block of L-type Ca2+ channels, whereas diltiazem weakly blocked HERG current (IC50=17.3 micromol/L), and nifedipine did not block HERG current. Verapamil 20-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 204-208 10325236-3 1999 In our experiments, verapamil caused high-affinity block of HERG current (IC50=143.0 nmol/L), a value close to those reported for verapamil block of L-type Ca2+ channels, whereas diltiazem weakly blocked HERG current (IC50=17.3 micromol/L), and nifedipine did not block HERG current. Verapamil 20-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 204-208 10325236-4 1999 Verapamil block of HERG channels was use and frequency dependent, and verapamil unbound from HERG channels at voltages near the normal cardiac cell resting potential or with drug washout. Verapamil 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 10325236-5 1999 Block of HERG current by verapamil was reduced by lowering pHO, which decreases the proportion of drug in the membrane-permeable neutral form. Verapamil 25-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 10325236-6 1999 N-methyl-verapamil, a membrane-impermeable, permanently charged verapamil analogue, blocked HERG channels only when applied intracellularly. n-methyl-verapamil 0-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 10325236-6 1999 N-methyl-verapamil, a membrane-impermeable, permanently charged verapamil analogue, blocked HERG channels only when applied intracellularly. Verapamil 9-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-96 10325236-7 1999 Verapamil antagonized dofetilide block of HERG channels, which suggests that they may share a common binding site. Verapamil 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 10325236-7 1999 Verapamil antagonized dofetilide block of HERG channels, which suggests that they may share a common binding site. dofetilide 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 10325236-10 1999 Thus, verapamil shares high-affinity HERG channel blocking properties with other class III antiarrhythmic drugs, and this may contribute to its antiarrhythmic mechanism. Verapamil 6-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 10369479-0 1999 Inhibition of HERG channels stably expressed in a mammalian cell line by the antianginal agent perhexiline maleate. perhexiline maleate 95-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 10369479-4 1999 Perhexiline caused voltage- and frequency-dependent block of HERG (IC50 7.8 microM). Perhexiline 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-65 10369479-6 1999 In conclusion, perhexiline potently inhibits transfected HERG channels and this is the probable mechanism for QT prolongation and torsades de pointes. Perhexiline 15-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 57-61 10187793-8 1999 The amino-terminal region of HERG was recently crystallized and shown to possess a Per-Arnt-Sim (PAS) domain. Aminosalicylic Acid 97-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 10187793-9 1999 The location of these mutations suggests they may disrupt the PAS domain and interfere with its interaction with the S4-S5 linker of the HERG channel. Aminosalicylic Acid 62-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 137-141 10376921-0 1999 Block of HERG potassium channels by the antihistamine astemizole and its metabolites desmethylastemizole and norastemizole. Astemizole 54-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 10376921-0 1999 Block of HERG potassium channels by the antihistamine astemizole and its metabolites desmethylastemizole and norastemizole. desmethylastemizole 85-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 10376921-0 1999 Block of HERG potassium channels by the antihistamine astemizole and its metabolites desmethylastemizole and norastemizole. tecastemizole 109-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 10376921-2 1999 Astemizole blocks the rapidly activating delayed rectifier K+ current I(Kr) and the human ether-a go-go-related gene (HERG) K+ channels that underlie it. Astemizole 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-122 10376921-6 1999 Our objective in the present work was to study the effects of desmethylastemizole, norastemizole, and astemizole on HERG K+ channels. desmethylastemizole 62-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 116-120 10376921-6 1999 Our objective in the present work was to study the effects of desmethylastemizole, norastemizole, and astemizole on HERG K+ channels. tecastemizole 83-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 116-120 10376921-6 1999 Our objective in the present work was to study the effects of desmethylastemizole, norastemizole, and astemizole on HERG K+ channels. Astemizole 71-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 116-120 10376921-8 1999 Desmethylastemizole and astemizole blocked HERG current with similar concentration dependence (half-maximal block of 1.0 and 0.9 nM, respectively) and block was use dependent. desmethylastemizole 0-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 10376921-8 1999 Desmethylastemizole and astemizole blocked HERG current with similar concentration dependence (half-maximal block of 1.0 and 0.9 nM, respectively) and block was use dependent. Astemizole 9-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 43-47 10376921-9 1999 Norastemizole also blocked HERG current; however, block was incomplete and required higher drug concentrations (half-maximal block of 27.7 nM). tecastemizole 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 10376921-10 1999 CONCLUSIONS: Desmethylastemizole and astemizole cause equipotent block of HERG channels, and these are among the most potent HERG channel antagonists yet studied. desmethylastemizole 13-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 10376921-10 1999 CONCLUSIONS: Desmethylastemizole and astemizole cause equipotent block of HERG channels, and these are among the most potent HERG channel antagonists yet studied. desmethylastemizole 13-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 10376921-10 1999 CONCLUSIONS: Desmethylastemizole and astemizole cause equipotent block of HERG channels, and these are among the most potent HERG channel antagonists yet studied. Astemizole 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 74-78 10376921-10 1999 CONCLUSIONS: Desmethylastemizole and astemizole cause equipotent block of HERG channels, and these are among the most potent HERG channel antagonists yet studied. Astemizole 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 125-129 10376921-12 1999 Norastemizole block of HERG channels is weaker; thus, the risk of producing ventricular arrhythmias may be lower. tecastemizole 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 10096894-1 1999 We have investigated actions of various divalent cations (Ba2+, Sr2+, Mn2+, Co2+, Ni2+, Zn2+) on human ether-a-go-go related gene (HERG) channels expressed in Xenopus laevis oocytes using the voltage clamp technique. strontium cation 64-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10096894-1 1999 We have investigated actions of various divalent cations (Ba2+, Sr2+, Mn2+, Co2+, Ni2+, Zn2+) on human ether-a-go-go related gene (HERG) channels expressed in Xenopus laevis oocytes using the voltage clamp technique. Manganese(2+) 70-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10096894-1 1999 We have investigated actions of various divalent cations (Ba2+, Sr2+, Mn2+, Co2+, Ni2+, Zn2+) on human ether-a-go-go related gene (HERG) channels expressed in Xenopus laevis oocytes using the voltage clamp technique. Cobalt(2+) 76-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10096894-1 1999 We have investigated actions of various divalent cations (Ba2+, Sr2+, Mn2+, Co2+, Ni2+, Zn2+) on human ether-a-go-go related gene (HERG) channels expressed in Xenopus laevis oocytes using the voltage clamp technique. Nickel(2+) 82-86 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10096894-1 1999 We have investigated actions of various divalent cations (Ba2+, Sr2+, Mn2+, Co2+, Ni2+, Zn2+) on human ether-a-go-go related gene (HERG) channels expressed in Xenopus laevis oocytes using the voltage clamp technique. Zinc 88-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 10086971-4 1999 METHODS AND RESULTS: New specific primers allowed the amplification of the 3" part of HERG, the identification of 2 missense mutations, S818L and V822 M, in the putative cyclic nucleotide binding domain, and a 1-bp insertion, 3108+1G. Nucleotides, Cyclic 170-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 10028924-0 1999 Modulation of HERG potassium channels by extracellular magnesium and quinidine. Magnesium 55-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 10208308-0 1999 Inhibitory effects of the class III antiarrhythmic drug amiodarone on cloned HERG potassium channels. Amiodarone 56-66 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 10208308-3 1999 In this study, we investigated the acute effects of the class III antiarrhythmic drug amiodarone on HERG channels expressed heterologously in Xenopus oocytes by use of the two-microelectrode voltage clamp technique. Amiodarone 86-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 10208308-4 1999 Amiodarone blocked HERG channels with an IC50 of 9.8 microM with a maximum outward tail current reduction of 62.8%. Amiodarone 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-23 10208308-7 1999 These results indicate that HERG channels can be blocked by amiodarone in closed, open and inactivated states. Amiodarone 60-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-32 10208308-9 1999 In summary, our data suggest that the strong class III antiarrhythmic action of amiodarone is at least partially based upon its acute inhibitory effects on HERG potassium channels. Amiodarone 80-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 156-160 9925876-0 1999 N-linked glycosylation sites determine HERG channel surface membrane expression. Nitrogen 0-1 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 9925876-4 1999 While a recent report indicates that LQT in some patients is associated with a mutation of HERG at a consensus extracellular N-linked glycosylation site (N629), earlier studies failed to identify a role for N-linked glycosylation in the functional expression of voltage-gated K+ channels. Nitrogen 125-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 9925876-4 1999 While a recent report indicates that LQT in some patients is associated with a mutation of HERG at a consensus extracellular N-linked glycosylation site (N629), earlier studies failed to identify a role for N-linked glycosylation in the functional expression of voltage-gated K+ channels. Nitrogen 154-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-95 9925876-5 1999 In this study we used pharmacological agents and site-directed mutagenesis to assess the contribution of N-linked glycosylation to the surface localization of HERG channels. Nitrogen 105-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 159-163 9925876-7 1999 Tunicamycin, an inhibitor of N-linked glycosylation, blocked normal surface membrane expression of a HERG-green fluorescent protein (GFP) fusion protein (HERGGFP) transiently expressed in human embryonic kidney (HEK 293) cells imaged with confocal microscopy. Tunicamycin 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 9925876-11 1999 Furthermore, patch clamp analysis revealed that there was a virtual absence of HERG current in the N-glycosylation mutants. Nitrogen 99-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 9925876-13 1999 Taken together, these results strongly suggest that N-linked glycosylation is required for surface membrane expression of HERG. Nitrogen 52-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-126 9925876-14 1999 These findings may provide insight into a mechanism responsible for LQT2 due to N-linked glycosylation-related mutations of HERG. Nitrogen 80-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 9925876-14 1999 These findings may provide insight into a mechanism responsible for LQT2 due to N-linked glycosylation-related mutations of HERG. Nitrogen 80-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 10028924-10 1999 Quinidine (3 microM) inhibited HERG currents expressed in oocytes by 32.1 +/- 3.2% (n = 5), whereas 1 microM quinidine inhibited HERG currents in tsA201 cells by 75.8 +/- 2.4% (n = 12). Quinidine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 31-35 10028924-10 1999 Quinidine (3 microM) inhibited HERG currents expressed in oocytes by 32.1 +/- 3.2% (n = 5), whereas 1 microM quinidine inhibited HERG currents in tsA201 cells by 75.8 +/- 2.4% (n = 12). Quinidine 109-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 129-133 10028924-0 1999 Modulation of HERG potassium channels by extracellular magnesium and quinidine. Quinidine 69-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 10028924-12 1999 These results indicate that extracellular Mg2+ exerts a direct action on HERG potassium channels, resulting in suppression of outward repolarizing potassium current. magnesium ion 42-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 10028924-14 1999 Potent suppression of HERG channel current by quinidine, compared with that of I(Ks) and I(Na), is a likely contributor to torsades de pointes arrhythmias. Quinidine 46-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 10028924-5 1999 This study was designed to investigate the effects of extracellular magnesium (Mg2+) on HERG potassium currents. Magnesium 68-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 10028924-5 1999 This study was designed to investigate the effects of extracellular magnesium (Mg2+) on HERG potassium currents. magnesium ion 79-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 10028924-5 1999 This study was designed to investigate the effects of extracellular magnesium (Mg2+) on HERG potassium currents. Potassium 93-102 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 10028924-7 1999 Extracellular Mg2+ (0.3-10 mM) caused a concentration-dependent shift in the membrane-potential dependence of HERG channel opening, causing a reduction in K+ current. magnesium ion 14-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 10688323-8 1999 Propranolol (1 micromol/L), a beta blocker, completely prevented the effect of isoproterenol to persistently or transiently increase TDR and to induce TdP in the LQT1 and LQT2 models, but facilitated TdP in the LQT3 model. Propranolol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 9882738-5 1999 Mutation of acidic residues (D540, E544) in the S4-S5 linker of HERG channels to neutral (Ala) or basic (Lys) residues accelerated the rate of channel deactivation. Alanine 90-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 9882738-5 1999 Mutation of acidic residues (D540, E544) in the S4-S5 linker of HERG channels to neutral (Ala) or basic (Lys) residues accelerated the rate of channel deactivation. Lysine 105-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 10090227-0 1999 Long-term (subacute) potassium treatment in congenital HERG-related long QT syndrome (LQTS2). Potassium 21-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-59 10688323-8 1999 Propranolol (1 micromol/L), a beta blocker, completely prevented the effect of isoproterenol to persistently or transiently increase TDR and to induce TdP in the LQT1 and LQT2 models, but facilitated TdP in the LQT3 model. Isoproterenol 79-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 9862440-0 1998 Sulfonylureas blockade of neural and cardiac HERG channels. Sulfonylurea Compounds 0-13 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 9862440-2 1998 In this paper we show that I(HERG), recorded in neuroblastoma cells and guinea-pig ventricular myocytes, was reversibly inhibited by the K(ATP) channel blocker glibenclamide (IC50 = 74 microM). Glyburide 160-173 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 9862440-4 1998 Another sulfonylurea, glimepiride, had less effective results in blocking I(HERG). Sulfonylurea Compounds 8-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 9862440-4 1998 Another sulfonylurea, glimepiride, had less effective results in blocking I(HERG). glimepiride 22-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 76-80 9679158-0 1998 Transfer of rapid inactivation and sensitivity to the class III antiarrhythmic drug E-4031 from HERG to M-eag channels. E 4031 84-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-100 9737994-0 1998 HERG potassium channel activation is shifted by phorbol esters via protein kinase A-dependent pathways. Phorbol Esters 48-62 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 9737994-11 1998 Forskolin (400 microM), an activator of the adenylate cyclase that results in PKA activation, shifted the HERG activation curve by 14 mV. Colforsin 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 9845367-0 1998 Crystal structure and functional analysis of the HERG potassium channel N terminus: a eukaryotic PAS domain. Protactinium 97-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-53 9765513-1 1998 We examined the effects of the calcium channel blockers nitrendipine, diltiazem, verapamil, bepridil, and mibefradil on the cloned HERG and KvLQT1/IsK K+ channels. Nitrendipine 56-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 9765513-1 1998 We examined the effects of the calcium channel blockers nitrendipine, diltiazem, verapamil, bepridil, and mibefradil on the cloned HERG and KvLQT1/IsK K+ channels. Diltiazem 70-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 9765513-1 1998 We examined the effects of the calcium channel blockers nitrendipine, diltiazem, verapamil, bepridil, and mibefradil on the cloned HERG and KvLQT1/IsK K+ channels. Verapamil 81-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 9765513-1 1998 We examined the effects of the calcium channel blockers nitrendipine, diltiazem, verapamil, bepridil, and mibefradil on the cloned HERG and KvLQT1/IsK K+ channels. Bepridil 92-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 9765513-1 1998 We examined the effects of the calcium channel blockers nitrendipine, diltiazem, verapamil, bepridil, and mibefradil on the cloned HERG and KvLQT1/IsK K+ channels. Mibefradil 106-116 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 9765513-3 1998 When expressed in transfected COS cells, HERG is blocked in a concentration-dependent manner by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM), and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Bepridil 96-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 9765513-3 1998 When expressed in transfected COS cells, HERG is blocked in a concentration-dependent manner by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM), and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Verapamil 127-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 9765513-3 1998 When expressed in transfected COS cells, HERG is blocked in a concentration-dependent manner by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM), and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Mibefradil 163-173 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 9765513-3 1998 When expressed in transfected COS cells, HERG is blocked in a concentration-dependent manner by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM), and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Nitrendipine 204-216 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 9765513-3 1998 When expressed in transfected COS cells, HERG is blocked in a concentration-dependent manner by bepridil (EC50 = 0.55 microM), verapamil (EC50 = 0.83 microM), and mibefradil (EC50 = 1.43 microM), whereas nitrendipine and diltiazem have negligible effects. Diltiazem 221-230 potassium voltage-gated channel subfamily H member 2 Homo sapiens 41-45 9679158-1 1998 The gating behaviour and pharmacological sensitivity of HERG are remarkably different from the corresponding properties of M-eag, a structurally similar member of the Eag family of potassium channels. m-eag 123-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 9679158-5 1998 Transfer of a small segment of the HERG polypeptide to M-eag, consisting largely of the P region and part of the S6 transmembrane domain, is sufficient to confer rapid inactivation and E-4031 sensitivity to M-eag. Methyl 2-acetamido-2-deoxy-beta-D-glucopyranoside 57-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 9714291-5 1998 In fact, interference with HERG K+ channels seems to be the main mechanism explaining both the therapeutic actions of the class III antiarrhythmics and the potential cardiotoxicity of second-generation H1 receptor antagonists such as terfenadine and astemizole, as well as of psychotropic drugs such as some antidepressants and neuroleptics. Terfenadine 234-245 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Terfenadine 292-303 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Erythromycin 307-319 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 9694935-0 1998 The antipsychotic agent sertindole is a high affinity antagonist of the human cardiac potassium channel HERG. sertindole 24-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 9694935-5 1998 Using patch clamp electrophysiology, we found sertindole blocked HERG currents with an IC50 value of 14.0 nM when tail currents at -40 mV were measured after a 2-sec depolarization to +20 mV. sertindole 46-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 9694935-11 1998 It is concluded that sertindole is a high affinity antagonist of the human cardiac potassium channel HERG and that this blockade underlies the prolongation of QT interval observed with this drug. sertindole 21-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 9694935-12 1998 Furthermore, the sertindole molecule may provide a useful starting point for the development of very high affinity ligands for HERG. sertindole 17-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 9694927-0 1998 Blockade of HERG and Kv1.5 by ketoconazole. Ketoconazole 30-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 9694927-3 1998 We previously showed that terfenadine blocked HERG (Human Ether-a-Gogo Related Gene), an important component of the repolarizing cardiac delayed rectifier IK with concentration needed to obtain 50% of the block (IC50) in the therapeutic range (300 nM). Terfenadine 26-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 9694927-5 1998 Whether Kv1.5 and HERG proteins are direct targets for ketoconazole has yet to be addressed. Ketoconazole 55-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 9694927-6 1998 We heterologously expressed HERG and Kv1.5 in Xenopus oocytes and compared their sensitivities to ketoconazole. Ketoconazole 98-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-32 9694927-11 1998 We conclude that ketoconazole may potentiate the effects of terfenadine first by an indirect pharmacokinetic action to elevate plasma levels and second by a direct pharmacodynamic action on HERG currents. Ketoconazole 17-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-194 9694927-11 1998 We conclude that ketoconazole may potentiate the effects of terfenadine first by an indirect pharmacokinetic action to elevate plasma levels and second by a direct pharmacodynamic action on HERG currents. Terfenadine 60-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-194 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Ketoconazole 100-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 9658196-0 1998 Molecular basis for the lack of HERG K+ channel block-related cardiotoxicity by the H1 receptor blocker cetirizine compared with other second-generation antihistamines. Cetirizine 104-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 9658196-1 1998 In the current study, the potential blocking ability of K+ channels encoded by the human ether-a-go-go related gene (HERG) by the piperazine H1 receptor antagonist cetirizine has been examined and compared with that of other second-generation antihistamines (astemizole, terfenadine, and loratadine). Cetirizine 164-174 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 9658196-1 1998 In the current study, the potential blocking ability of K+ channels encoded by the human ether-a-go-go related gene (HERG) by the piperazine H1 receptor antagonist cetirizine has been examined and compared with that of other second-generation antihistamines (astemizole, terfenadine, and loratadine). Astemizole 259-269 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 9658196-1 1998 In the current study, the potential blocking ability of K+ channels encoded by the human ether-a-go-go related gene (HERG) by the piperazine H1 receptor antagonist cetirizine has been examined and compared with that of other second-generation antihistamines (astemizole, terfenadine, and loratadine). Terfenadine 271-282 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 9658196-1 1998 In the current study, the potential blocking ability of K+ channels encoded by the human ether-a-go-go related gene (HERG) by the piperazine H1 receptor antagonist cetirizine has been examined and compared with that of other second-generation antihistamines (astemizole, terfenadine, and loratadine). Loratadine 288-298 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 9658196-3 1998 On the other hand, terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (the estimated IC50 values were 330 and 480 nM, respectively), whereas loratadine was approximately 300-fold less potent (IC50 approximately 100 microM). Terfenadine 19-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 9658196-3 1998 On the other hand, terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (the estimated IC50 values were 330 and 480 nM, respectively), whereas loratadine was approximately 300-fold less potent (IC50 approximately 100 microM). Astemizole 35-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 9658196-8 1998 Furthermore, the application of cetirizine (3 microM) on the intracellular side of the membrane of HERG-transfected human embryonic kidney 293 cells did not affect IHERG, whereas the same intracellular concentration of astemizole caused a complete block. Cetirizine 32-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-103 9658196-10 1998 Cetirizine in particular, which possesses more polar and smaller substituent groups attached to the tertiary amine compared with other antihistamines, lacks HERG-blocking properties, possibly explaining the absence of torsade de pointes ventricular arrhythmias associated with its therapeutical use. Cetirizine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 157-161 9714291-5 1998 In fact, interference with HERG K+ channels seems to be the main mechanism explaining both the therapeutic actions of the class III antiarrhythmics and the potential cardiotoxicity of second-generation H1 receptor antagonists such as terfenadine and astemizole, as well as of psychotropic drugs such as some antidepressants and neuroleptics. Astemizole 250-260 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 9654228-1 1998 INTRODUCTION: Inherited long QT syndrome (LQTS) recently has been associated with mutations in genes coding for potassium (KVLQT1, KCNE1, and HERG) or sodium (SCN5A) ion channels involved in regulating either sodium inward or potassium outward currents of heart cells, resulting in prolongation of the repolarization period. Potassium 112-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 9654228-1 1998 INTRODUCTION: Inherited long QT syndrome (LQTS) recently has been associated with mutations in genes coding for potassium (KVLQT1, KCNE1, and HERG) or sodium (SCN5A) ion channels involved in regulating either sodium inward or potassium outward currents of heart cells, resulting in prolongation of the repolarization period. Sodium 209-215 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 9654228-1 1998 INTRODUCTION: Inherited long QT syndrome (LQTS) recently has been associated with mutations in genes coding for potassium (KVLQT1, KCNE1, and HERG) or sodium (SCN5A) ion channels involved in regulating either sodium inward or potassium outward currents of heart cells, resulting in prolongation of the repolarization period. Potassium 226-235 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 9570196-3 1998 Genes responsible for LQT1, LQT2, and LQT3 have been identified as cardiac potassium channel genes (KVLQT1, HERG) and the cardiac sodium channel gene (SCN5A). Sodium 130-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 28-32 9535729-0 1998 Long-term exposure to retinoic acid induces the expression of IRK1 channels in HERG channel-endowed neuroblastoma cells. Tretinoin 22-35 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-83 9490815-13 1998 Down-regulation of h-eag by long-term exposure to retinoic acid was paralleled by a right shift in the activation potential of HERG-like channels. Tretinoin 50-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 9535729-3 1998 After 10-20 days exposure to Retinoic Acid (RA), SH-SY5Y cells showed, in addition to HERG currents, a novel current characterized by inward rectification, dependence on the extracellular K+ concentration, and blockade by Cs+ and Ba2+, the main features of the IRK1 current. Tretinoin 29-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 9535729-3 1998 After 10-20 days exposure to Retinoic Acid (RA), SH-SY5Y cells showed, in addition to HERG currents, a novel current characterized by inward rectification, dependence on the extracellular K+ concentration, and blockade by Cs+ and Ba2+, the main features of the IRK1 current. Tretinoin 44-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 9535729-6 1998 On the whole, data here presented demonstrate that RA-induced neuronal differentiation of neuroblastoma cells is accompanied by the switch from a HERG-driven to a IRK1-driven control of Vrest, similarly to what happens in normal differentiating neurons; however, in tumor cells, this switch does not imply the abolition of HERG channel expression. Tretinoin 51-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 146-150 9535729-6 1998 On the whole, data here presented demonstrate that RA-induced neuronal differentiation of neuroblastoma cells is accompanied by the switch from a HERG-driven to a IRK1-driven control of Vrest, similarly to what happens in normal differentiating neurons; however, in tumor cells, this switch does not imply the abolition of HERG channel expression. Tretinoin 51-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 323-327 9508824-0 1998 Voltage-dependent blockade of HERG channels expressed in Xenopus oocytes by external Ca2+ and Mg2+. magnesium ion 94-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 9508824-3 1998 Using a two-microelectrode voltage clamp technique, the effect of external Ca2+ and Mg2+ on the HERG current (IHERG) was investigated. magnesium ion 84-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 96-100 9508824-18 1998 These results suggest that external Ca2+ and Mg2+ block the HERG channel in a voltage- and time-dependent manner, resulting in a voltage dependence which has been regarded as a property of the activation gate. magnesium ion 45-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-64 9484850-0 1998 Blockade of HERG channels by the class III antiarrhythmic azimilide: mode of action. azimilide 58-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 9556090-8 1998 Raising the serum potassium concentration can increase outward HERG potassium current and is effective in shortening the QTc of patients with HERG mutations. Potassium 18-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 9556090-8 1998 Raising the serum potassium concentration can increase outward HERG potassium current and is effective in shortening the QTc of patients with HERG mutations. Potassium 18-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-146 9556090-8 1998 Raising the serum potassium concentration can increase outward HERG potassium current and is effective in shortening the QTc of patients with HERG mutations. Potassium 68-77 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 9484850-10 1998 Azimilide blockade of HERG channels expressed in Xenopus oocytes and I(Kr) in mouse AT-1 cells was decreased under conditions of high [K+]e, whereas block of slowly activating I(Ks) channels was not affected by changes in [K+]e. 5. azimilide 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 22-26 9484850-11 1998 In summary, azimilide is a blocker of cardiac delayed rectifier channels, I(Ks) and HERG. azimilide 12-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 9486667-0 1998 Molecular determinants of dofetilide block of HERG K+ channels. dofetilide 26-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 9486667-2 1998 HERG/IKr channels are a prime target for the pharmacological management of arrhythmias and are selectively blocked by class III antiarrhythmic methanesulfonanilide drugs, such as dofetilide, E4031, and MK-499, at submicromolar concentrations. N-Phenylmethanesulfonamide 143-163 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 9486667-2 1998 HERG/IKr channels are a prime target for the pharmacological management of arrhythmias and are selectively blocked by class III antiarrhythmic methanesulfonanilide drugs, such as dofetilide, E4031, and MK-499, at submicromolar concentrations. dofetilide 179-189 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 9486667-2 1998 HERG/IKr channels are a prime target for the pharmacological management of arrhythmias and are selectively blocked by class III antiarrhythmic methanesulfonanilide drugs, such as dofetilide, E4031, and MK-499, at submicromolar concentrations. E 4031 191-196 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 9486667-2 1998 HERG/IKr channels are a prime target for the pharmacological management of arrhythmias and are selectively blocked by class III antiarrhythmic methanesulfonanilide drugs, such as dofetilide, E4031, and MK-499, at submicromolar concentrations. L 706000 202-208 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 9486667-5 1998 Using constructs heterologously expressed in Xenopus oocytes, we found that transplantation of the S5-S6 linker from BEAG into HERG removed high-affinity block by dofetilide. dofetilide 163-173 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 9486667-7 1998 Thus, our results indicate that important determinants of dofetilide binding are localized to the pore region of HERG. dofetilide 58-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 113-117 9486667-11 1998 Thus, the serine in position HERG 620 may participate directly in dofetilide binding; however, an intact C-type inactivation process seems to be crucial for high-affinity drug binding. Serine 10-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 9486667-11 1998 Thus, the serine in position HERG 620 may participate directly in dofetilide binding; however, an intact C-type inactivation process seems to be crucial for high-affinity drug binding. dofetilide 66-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 9484850-3 1998 Because HERG channels underly the conductance I(Kr), in human heart, the effects of azimilide on HERG channels expressed in Xenopus oocytes were the focus of the present study. azimilide 84-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 9484850-5 1998 In contrast to other well characterized HERG channel blockers, azimilide blockade was reverse use-dependent, i.e., the relative block and apparent affinity of azimilide decreased with an increase in channel activation frequency. azimilide 63-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 9484850-6 1998 Azimilide blocked HERG channels at 0.1 and 1 Hz with IC50s of 1.4 microM and 5.2 microM respectively. azimilide 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 9484850-8 1998 In an envelope of tail test, HERG channel blockade increased with increasing channel activation, indicating binding of azimilide to open channels. azimilide 119-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 9359911-9 1997 Furthermore, the inwardly rectifying current was blocked by astemizole, a potent and selective inhibitor of human ether-a-go-go -related gene (HERG) K+ channels. Astemizole 60-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 143-147 9449325-12 1998 HERG current was blocked by low concentrations of E-4031 (IC50 7.7 nM), a value close to that reported for I(Kr) in native cardiac myocytes. E 4031 50-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 9517465-4 1997 This current (quail I[IR] or ql[IR]), which is active at membrane potentials positive to -35 mV, was blocked by Cs+ and by class III antiarrhythmic drugs, thus resembling the K+ current encoded by the human ether-a-go-go-related gene (HERG). Cesium 112-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 235-239 9395068-0 1997 A mechanism for the proarrhythmic effects of cisapride (Propulsid): high affinity blockade of the human cardiac potassium channel HERG. Cisapride 45-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 9395068-0 1997 A mechanism for the proarrhythmic effects of cisapride (Propulsid): high affinity blockade of the human cardiac potassium channel HERG. Cisapride 56-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 130-134 9395068-5 1997 Using patch clamp electrophysiology, we found that cisapride was a potent inhibitor of HERG displaying an IC50 value of 44.5 nmol/l when tail currents at -40 mV were measured following a 2 s test depolarization to +20 mV. Cisapride 51-60 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-91 9395068-6 1997 When HERG currents were measured at the end of prolonged (20 s) depolarizing steps to +20 mV, the apparent affinity of cisapride was increased and measured 6.70 nmol/l. Cisapride 119-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 5-9 9395068-7 1997 The main effect of cisapride was to enhance the rate of HERG current decay thereby reducing current at the end of the voltage clamp pulse. Cisapride 19-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 9395068-8 1997 Furthermore, the potency of cisapride for the HERG channel was similar to that observed for the class III antiarrhythmic agent dofetilide (IC50 = 15.3 nmol/l) and the nonsedating antihistamine terfenadine (IC50 = 56.0 nmol/l). Cisapride 28-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 9395068-10 1997 It is concluded that cisapride displays specific, high affinity block of the human cardiac K+ channel HERG. Cisapride 21-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-106 9351462-7 1997 The Merg1 isoforms, like HERG, produce inwardly rectifying E-4031-sensitive currents when heterologously expressed in Xenopus oocytes. E 4031 59-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 9374794-0 1997 Blockage of the HERG human cardiac K+ channel by the gastrointestinal prokinetic agent cisapride. Cisapride 87-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 16-20 9374794-3 1997 The human ether-a-go-go-related gene (HERG), which encodes the rapidly activating delayed rectifier K+ current and is important in cardiac repolarization, may serve as a target for the action of cisapride. Cisapride 195-204 potassium voltage-gated channel subfamily H member 2 Homo sapiens 38-42 9374794-4 1997 We tested the hypothesis that cisapride blocks HERG. Cisapride 30-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-51 9374794-6 1997 Under voltage-clamp conditions, cisapride block of HERG is dose dependent with a half-maximal inhibitory concentration of 6.5 nM at 22 degrees C (n = 25 cells). Cisapride 32-41 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 9374794-9 1997 Block of HERG with cisapride after channel activation was voltage dependent. Cisapride 19-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 9374794-10 1997 At -20 mV, 10 nM cisapride reduced HERG tail-current amplitude by 5%, whereas, at + 20 mV, the tail-current amplitude was reduced by 45% (n = 4 cells). Cisapride 17-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-39 9374794-12 1997 We conclude that cisapride is a potent blocker of HERG channels expressed in HEK293 cells. Cisapride 17-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 9326673-0 1997 Regulation of the human ether-a-gogo related gene (HERG) K+ channels by reactive oxygen species. Reactive Oxygen Species 72-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-55 9395171-1 1997 INTRODUCTION: Recent clinical studies have reported a greater effectiveness of sodium channel block with mexiletine to abbreviate the QT interval in patients with the chromosome 3 variant (SCN5A, LQT3) of the long QT syndrome (LQTS) than those with the chromosome 7 form of the disease (HERG, LQT2), suggesting the possibility of gene-specific therapy for the two distinct forms of the congenital LQTS. Mexiletine 105-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 287-291 9395171-1 1997 INTRODUCTION: Recent clinical studies have reported a greater effectiveness of sodium channel block with mexiletine to abbreviate the QT interval in patients with the chromosome 3 variant (SCN5A, LQT3) of the long QT syndrome (LQTS) than those with the chromosome 7 form of the disease (HERG, LQT2), suggesting the possibility of gene-specific therapy for the two distinct forms of the congenital LQTS. Mexiletine 105-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 293-297 9395171-14 1997 The data provide further support for the hypothesis that block of the late sodium current may be of value in the treatment of LQT2 as well as LQT3 and perhaps other congenital and acquired (drug-induced) forms of LQTS. Sodium 75-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 126-130 9326673-2 1997 For this reason, the possible modulation by reactive oxygen species (ROS) of HERG and other cloned K+ channels expressed in Xenopus oocytes has been explored in the present study. Reactive Oxygen Species 44-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 9326673-2 1997 For this reason, the possible modulation by reactive oxygen species (ROS) of HERG and other cloned K+ channels expressed in Xenopus oocytes has been explored in the present study. Reactive Oxygen Species 69-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 9326673-5 1997 This ROS-induced increase in HERG outward K+ currents was due to a depolarizing shift of the voltage-dependence of channel inactivation, with no change in channel activation. Reactive Oxygen Species 5-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-33 9326673-7 1997 Fe/Asc-induced stimulation of HERG outward currents was completely prevented by perfusion of the oocytes with a ROS scavenger mixture (containing 1,000 units/ml catalase, 200 ng/ml superoxide dismutase, and 2 mM mannitol). Reactive Oxygen Species 112-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 9326673-7 1997 Fe/Asc-induced stimulation of HERG outward currents was completely prevented by perfusion of the oocytes with a ROS scavenger mixture (containing 1,000 units/ml catalase, 200 ng/ml superoxide dismutase, and 2 mM mannitol). Mannitol 212-220 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-34 9326673-9 1997 In conclusion, the present results seem to suggest that changes in ROS production can specifically influence K+ currents carried by the HERG channels. Reactive Oxygen Species 67-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 136-140 9272507-11 1997 Conversely, LQT2 patients are at higher risk to develop syncope under stressful conditions, because of the combined arrhythmogenic effect of catecholamines with the insufficient adaptation of their QT interval. Catecholamines 141-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 9315735-0 1997 Specific block of cloned Herg channels by clofilium and its tertiary analog LY97241. clofilium 42-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 9315735-0 1997 Specific block of cloned Herg channels by clofilium and its tertiary analog LY97241. LY 97241 76-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 9315735-2 1997 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 52-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 167-171 9315735-2 1997 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). LY 97241 86-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 167-171 9315735-3 1997 Clofilium blocked HERG channels in a voltage-dependent fashion with an IC50 of 250 nM and 150 nM at 0 and +40 mV, respectively. clofilium 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 9315735-6 1997 Block of HERG channels by LY97241 was voltage dependent and the rate of HERG inactivation was increased by LY97241. LY 97241 26-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 9315735-6 1997 Block of HERG channels by LY97241 was voltage dependent and the rate of HERG inactivation was increased by LY97241. LY 97241 26-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 9315735-6 1997 Block of HERG channels by LY97241 was voltage dependent and the rate of HERG inactivation was increased by LY97241. LY 97241 107-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-13 9315735-6 1997 Block of HERG channels by LY97241 was voltage dependent and the rate of HERG inactivation was increased by LY97241. LY 97241 107-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-76 9315735-8 1997 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. LY 97241 136-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 9315735-8 1997 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. LY 97241 170-177 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 9315735-8 1997 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. LY 97241 170-177 potassium voltage-gated channel subfamily H member 2 Homo sapiens 201-205 9315735-9 1997 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. clofilium 41-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 146-150 9315735-9 1997 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. LY 97241 66-73 potassium voltage-gated channel subfamily H member 2 Homo sapiens 146-150 9272507-12 1997 Along the same line of development of gene-specific therapy, recent data demonstrated that an increase in the extracellular concentration of potassium shortens the QT interval in LQT2 patients suggesting that intervention aimed at increasing potassium plasma levels may represent a specific treatment for LQT2. Potassium 141-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 9272507-12 1997 Along the same line of development of gene-specific therapy, recent data demonstrated that an increase in the extracellular concentration of potassium shortens the QT interval in LQT2 patients suggesting that intervention aimed at increasing potassium plasma levels may represent a specific treatment for LQT2. Potassium 141-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 305-309 9272507-12 1997 Along the same line of development of gene-specific therapy, recent data demonstrated that an increase in the extracellular concentration of potassium shortens the QT interval in LQT2 patients suggesting that intervention aimed at increasing potassium plasma levels may represent a specific treatment for LQT2. Potassium 242-251 potassium voltage-gated channel subfamily H member 2 Homo sapiens 179-183 9272507-12 1997 Along the same line of development of gene-specific therapy, recent data demonstrated that an increase in the extracellular concentration of potassium shortens the QT interval in LQT2 patients suggesting that intervention aimed at increasing potassium plasma levels may represent a specific treatment for LQT2. Potassium 242-251 potassium voltage-gated channel subfamily H member 2 Homo sapiens 305-309 9230439-0 1997 A minK-HERG complex regulates the cardiac potassium current I(Kr). Potassium 42-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 7-11 9138706-0 1997 The inhibitory effect of the antipsychotic drug haloperidol on HERG potassium channels expressed in Xenopus oocytes. Haloperidol 48-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 9297927-4 1997 Both LQT1 and LQT2 relate with inward rectifying potassium currents and is repolarization related, therefore, it is speculate that patients of LQT1 and LQT2 may have an abnormal T-U-wave on their surface ECG. Potassium 49-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 9297927-4 1997 Both LQT1 and LQT2 relate with inward rectifying potassium currents and is repolarization related, therefore, it is speculate that patients of LQT1 and LQT2 may have an abnormal T-U-wave on their surface ECG. Potassium 49-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 152-156 9138706-3 1997 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. Haloperidol 173-184 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-116 9138706-7 1997 In contrast, haloperidol blocked HERG channels potently with an IC50 value of approximately 1 microM. Haloperidol 13-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 33-37 9138706-10 1997 Haloperidol block was use- and voltage-dependent, suggesting that it binds preferentially to either open or inactivated HERG channels. Haloperidol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 120-124 9138706-11 1997 As haloperidol increased the degree and rate of HERG inactivation, binding to inactivated HERG channels is suggested. Haloperidol 3-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 9138706-11 1997 As haloperidol increased the degree and rate of HERG inactivation, binding to inactivated HERG channels is suggested. Haloperidol 3-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 9138706-14 1997 Haloperidol block of HERG S631A at 0 mV was four fold weaker than for HERG wild-type channels. Haloperidol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-25 9138706-15 1997 Haloperidol affinity for HERG S631A was increased four fold at +40 mV compared to 0 mV. Haloperidol 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 25-29 9138706-17 1997 In summary, the data suggest that HERG channel blockade is involved in the arrhythmogenic side effects of haloperidol. Haloperidol 106-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 34-38 9138706-18 1997 The mechanism of haloperidol block involves binding to inactivated HERG channels. Haloperidol 17-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 8995352-5 1997 Truncated HERG proteins containing NAB(HERG), including one that resulted from the delta1261 human mutation, inhibit the functional expression of the HERG channel in transfected cells. nab 35-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 10-14 9087606-7 1997 Single HERG channels were blocked by MK-499, a class III antiarrhythmic agent that blocks I(Kr) in cardiac myocytes. L 706000 37-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 7-11 8995352-5 1997 Truncated HERG proteins containing NAB(HERG), including one that resulted from the delta1261 human mutation, inhibit the functional expression of the HERG channel in transfected cells. nab 35-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 8995352-5 1997 Truncated HERG proteins containing NAB(HERG), including one that resulted from the delta1261 human mutation, inhibit the functional expression of the HERG channel in transfected cells. nab 35-38 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 8873679-8 1996 The SCN5A mutations result in defective sodium channel inactivation, whereas HERG mutations result in decreased outward potassium current. Potassium 120-129 potassium voltage-gated channel subfamily H member 2 Homo sapiens 77-81 8921803-10 1996 In excised macro patches, HERG currents were blocked by the class III antiarrhythmic drug dofetilide, with an IC50 of 35 nmol/L. dofetilide 90-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 8921803-11 1996 Dofetilide block was slow and greatly attenuated at positive potentials at which HERG rectifies. dofetilide 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 9117354-3 1996 IIR was blocked by Cs+ ions and by the antiarrhythmic drug E-4031, a specific inhibitor of the HERG-codified channels. E 4031 59-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 95-99 8641472-0 1996 Blockade of HERG channels expressed in Xenopus oocytes by the histamine receptor antagonists terfenadine and astemizole. Terfenadine 93-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 8952843-5 1996 We developed a cellular model in which ventricular myocytes were exposed to anthopleurin and dofetilide in order to mimic LQT3 and LQT2, respectively. anthopleurin 76-88 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 8952843-5 1996 We developed a cellular model in which ventricular myocytes were exposed to anthopleurin and dofetilide in order to mimic LQT3 and LQT2, respectively. dofetilide 93-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 131-135 8952843-11 1996 Conversely, LQT2 patients are at higher risk to develop syncope under stressful conditions, because of the combined arrhythmogenic effect of cathecolamines with the insufficient adaptation of their QT interval when heart rate increases. cathecolamines 141-155 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 8790040-2 1996 BACKGROUND: Many members of families with inherited long-QT (LQT) syndrome have mutations in HERG, a gene encoding a cardiac potassium channel that is modulated by extracellular potassium. Potassium 125-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-97 8799887-12 1996 Activity of HERG channels depended on extracellular cations, which are effective for channel activation, in the order Cs+ > K+ > > Li+ > Na+. Cesium 118-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 8649354-0 1996 High affinity open channel block by dofetilide of HERG expressed in a human cell line. dofetilide 36-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 50-54 8635257-4 1996 A single nucleotide substitution of thymidine to guanine (T1961G) changed the coding sense of HERG from isoleucine to arginine (Ile593Arg) in the channel pore region. Thymidine 36-45 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 8635257-4 1996 A single nucleotide substitution of thymidine to guanine (T1961G) changed the coding sense of HERG from isoleucine to arginine (Ile593Arg) in the channel pore region. Guanine 49-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 8914737-0 1996 Novel missense mutation in the cyclic nucleotide-binding domain of HERG causes long QT syndrome. Nucleotides, Cyclic 31-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-71 8914737-11 1996 Furthermore, the location and character of this mutation suggests that the cyclic nucleotide-binding domain of the potassium channel encoded by HERG plays an important role in normal cardiac repolarization and may decrease susceptibility to ventricular tachyarrhythmias. Nucleotides, Cyclic 75-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 144-148 8772706-0 1996 HERG, a primary human ventricular target of the nonsedating antihistamine terfenadine. antihistamine terfenadine 60-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 8772706-5 1996 We found that HERG was 10 times more sensitive than Kv1.5 to terfenadine block. Terfenadine 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 8772706-8 1996 The Kd value for HERG block is relevant to the toxicity of the antihistamine, since the clinical terfenadine concentrations in human plasma may reach the 100 nmol/L range. Terfenadine 97-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 8772706-10 1996 We propose that the blocking of HERG by terfenadine explains the acquired long QT syndrome. Terfenadine 40-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 8877771-4 1996 By SSCP analysis and direct sequencing, we determined a new missense mutation in the HERG coding sequence, a G to A transition at position 1681 resulting in the substitution of threonine for a highly conserved alanine at codon 561. Threonine 177-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 8877771-4 1996 By SSCP analysis and direct sequencing, we determined a new missense mutation in the HERG coding sequence, a G to A transition at position 1681 resulting in the substitution of threonine for a highly conserved alanine at codon 561. Alanine 210-217 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-89 8766823-3 1996 We measured the instantaneous current to voltage relationship for h-erg channels using the saponin permeabilized variation of the cut-open oocyte clamp technique. Saponins 91-98 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-71 8635257-4 1996 A single nucleotide substitution of thymidine to guanine (T1961G) changed the coding sense of HERG from isoleucine to arginine (Ile593Arg) in the channel pore region. Isoleucine 104-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 8635257-4 1996 A single nucleotide substitution of thymidine to guanine (T1961G) changed the coding sense of HERG from isoleucine to arginine (Ile593Arg) in the channel pore region. Arginine 118-126 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 8635257-4 1996 A single nucleotide substitution of thymidine to guanine (T1961G) changed the coding sense of HERG from isoleucine to arginine (Ile593Arg) in the channel pore region. ile593arg 128-137 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-98 8635257-6 1996 CONCLUSIONS: We conclude that the Ile593Arg missense mutation in HERG is the cause of LQT in this family because it segregates with disease, its presence was confirmed in three ways, and it is not found in normal individuals. ile593arg 34-43 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 8635257-7 1996 The Ile593Arg mutation may result in a change in potassium selectivity and permeability leading to a loss of HERG function, thereby resulting in LQT. Potassium 49-58 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 8641472-0 1996 Blockade of HERG channels expressed in Xenopus oocytes by the histamine receptor antagonists terfenadine and astemizole. Astemizole 109-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 8641472-2 1996 Here, terfenadine and astemizole both inhibited the human ether-a-go-go related gene (HERG) encoded channels expressed in Xenopus oocytes at nanomolar concentrations in a use- and voltage-dependent fashion. Terfenadine 6-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 8641472-2 1996 Here, terfenadine and astemizole both inhibited the human ether-a-go-go related gene (HERG) encoded channels expressed in Xenopus oocytes at nanomolar concentrations in a use- and voltage-dependent fashion. Astemizole 22-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 8641472-4 1996 These results suggest that blockade of HERG channels by terfenadine and astemizole might contribute to the cardiac side effects of these compounds. Terfenadine 56-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 8641472-4 1996 These results suggest that blockade of HERG channels by terfenadine and astemizole might contribute to the cardiac side effects of these compounds. Astemizole 72-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 8593709-8 1996 However, MK-499 did block HERG current if oocytes were repetitively pulsed, or clamped at a voltage positive to the threshold potential for channel activation. L 706000 9-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-30 8593709-11 1996 Under steady state conditions, block of HERG by MK-499 was half maximal at 123 +/- 12 nmol/L at a test potential of -20 mV. L 706000 48-54 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 34592322-3 2021 Previously we reported BZT and DMP as high-affinity human ether-a-go-go related gene (HERG) channel inhibitors with unknown proarrhythmic risk. Benzethonium 23-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 7604285-2 1995 The properties of HERG channels are consistent with the gating properties of Eag-related and other outwardly rectifying, S4-containing potassium channels, but with the addition of an inactivation mechanism that attenuates potassium efflux during depolarization. Potassium 135-144 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-22 24356123-8 2013 HERG channel phosphorylation was assayed by [(32)P]orthophosphate methods. [(32)p]orthophosphate 44-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-4 24356123-11 2013 Antioxidants vitamin E and MnTBAP both abolished the depressive effects of PKA or PKC activators on HERG function. Vitamin E 13-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 24356123-13 2013 CONCLUSIONS: HERG function is insensitive to PKA or PKC phosphorylation modulation per se, but can be impaired by the activators of PKA or PKC with long exposure likely via generation of ROS. Reactive Oxygen Species 187-190 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-17 24356123-14 2013 In view of the critical role of HERG K(+) channel in regulating cardiac repolarization and the sustained activation of both PKA and PKC in many pathological conditions of the heart such as heart failure, it is conceivable that HERG impairment by ROS accumulation induced by PKA and PKC contributes to the impaired cardiac repolarization. Reactive Oxygen Species 246-249 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 24356123-14 2013 In view of the critical role of HERG K(+) channel in regulating cardiac repolarization and the sustained activation of both PKA and PKC in many pathological conditions of the heart such as heart failure, it is conceivable that HERG impairment by ROS accumulation induced by PKA and PKC contributes to the impaired cardiac repolarization. Reactive Oxygen Species 246-249 potassium voltage-gated channel subfamily H member 2 Homo sapiens 227-231 34767653-1 2022 BACKGROUND: The T. indotineae population shows a high amount of terbinafine resistant isolates based on different point mutations of squalene epoxidase erg1 (ergosterol) gene. Terbinafine 64-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 152-156 34727194-3 2021 This study was undertaken to investigate the ability of phenanthrene to interact with hERG (human Ether-a-go-go-Related Gene) encoded Kv11.1 K+ channels, which play a central role in human ventricular repolarization. phenanthrene 56-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 134-140 8521555-4 1995 We tested the hypothesis that the QT interval would shorten more in LQT3 than in LQT2 patients in response to mexiletine and also in response to increases in heart rate. Mexiletine 110-120 potassium voltage-gated channel subfamily H member 2 Homo sapiens 81-85 8521555-12 1995 Conversely, LQT2 patients may be at higher risk to develop syncope under stressful conditions because of the combined arrhythmogenic effect of catecholamines with the insufficient adaptation of their QT interval when heart rate increases. Catecholamines 143-157 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 34907251-14 2021 Of note, dexmedetomidine might be antiarrhythmic in acquired LQT2 by reducing SDR. Dexmedetomidine 9-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-65 34592322-3 2021 Previously we reported BZT and DMP as high-affinity human ether-a-go-go related gene (HERG) channel inhibitors with unknown proarrhythmic risk. domiphen 31-34 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 34427958-8 2021 Sodium channel blockers significantly shortened QTc both in LQT3 and LQT2 patients, while the QTc shortening effect in LQT3 was superior to that in LQT2 (57.39 ms vs. 36.61 ms). Sodium 0-6 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-73 34427958-10 2021 CONCLUSIONS: sodium channel blockers can be useful both in LQT3 and LQT2 patients. Sodium 13-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 35455047-0 2022 KCNH2 p.Gly262AlafsTer98: A New Threatening Variant Associated with Long QT Syndrome in a Spanish Cohort. gly262alafster98 8-24 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-5 34383081-4 2022 The observed spontaneous inhibition of outward currents persisted in the presence of 4-aminopyridine, tetraethylammonium chloride or E-4031, the selective class III antiarrhythmic agent that blocks HERG channels. 4-Aminopyridine 85-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 198-202 34383081-4 2022 The observed spontaneous inhibition of outward currents persisted in the presence of 4-aminopyridine, tetraethylammonium chloride or E-4031, the selective class III antiarrhythmic agent that blocks HERG channels. Tetraethylammonium 102-129 potassium voltage-gated channel subfamily H member 2 Homo sapiens 198-202 34383081-4 2022 The observed spontaneous inhibition of outward currents persisted in the presence of 4-aminopyridine, tetraethylammonium chloride or E-4031, the selective class III antiarrhythmic agent that blocks HERG channels. E 4031 133-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 198-202 34901807-1 2021 A novel frameshift mutation in the KCNH2 gene for long QT syndrome type 2 (LQTS2) was identified after torsades des pointes ventricular tachycardia in a 49-year-old patient managed with octreotide and nadolol for an acute variceal bleed. Octreotide 186-196 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-40 34901807-1 2021 A novel frameshift mutation in the KCNH2 gene for long QT syndrome type 2 (LQTS2) was identified after torsades des pointes ventricular tachycardia in a 49-year-old patient managed with octreotide and nadolol for an acute variceal bleed. Nadolol 201-208 potassium voltage-gated channel subfamily H member 2 Homo sapiens 35-40 35587358-3 2022 Our data uncover a key enzyme of sterol synthesis, the hairpin membrane protein squalene monooxygenase (Erg1), as a non-canonical GET pathway client, thus rationalizing the lipotoxicity phenotypes of GET pathway mutants. Sterols 33-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 104-108 35587358-5 2022 Intriguingly, we find that the GET pathway is especially important for the acute upregulation of Erg1 induced by low sterol conditions. Sterols 117-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-101 34514026-7 2021 Consequently, human ether-a-go-go-related gene (HERG) channel blockers with high arrhythmogenic risk caused prolongation of contraction-relaxation duration and arrhythmia-like waveforms. ether-a 20-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 48-52 35525425-7 2022 In LQT2 women, linear mixed effects models showed significant inverse correlations of QTc with progesterone (p<0.001), and the progesterone to estradiol ratio (p<0.001). Progesterone 95-107 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 35525425-7 2022 In LQT2 women, linear mixed effects models showed significant inverse correlations of QTc with progesterone (p<0.001), and the progesterone to estradiol ratio (p<0.001). Progesterone 127-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 35525425-7 2022 In LQT2 women, linear mixed effects models showed significant inverse correlations of QTc with progesterone (p<0.001), and the progesterone to estradiol ratio (p<0.001). Estradiol 143-152 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 35525425-8 2022 Inverse relationships of the RR interval with estradiol levels (p=0.003) and of the T-wave duration with testosterone levels (p=0.014) were also observed in women with LQT2. Testosterone 105-117 potassium voltage-gated channel subfamily H member 2 Homo sapiens 168-172 35455047-4 2022 In this study, we identified all index patients referred for NGS genetic sequencing due to LQTS, in a Spanish cohort, who were carriers of a new pathogenic variant (KCNH2 p.Gly262AlafsTer98). gly262alafster98 173-189 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-170 35125346-4 2022 Methods: The thallium (Tl+) flux assay technique, widely used for drug screening, was optimized using human embryonic kidney (HEK-293) cells expressing a trafficking-deficient KV11.1 variant in 384-well plates. Thallium 13-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 176-182 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. Thallium 196-199 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-97 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. VU0405601 158-167 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-97 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. Thallium 196-199 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-138 35125346-8 2022 To validate the optimized trafficking assay, we carried out a pilot screen that identified three drugs (ibutilide, azaperone, and azelastine) as drugs that increase KV11.1 trafficking. ibutilide 104-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-171 35125346-8 2022 To validate the optimized trafficking assay, we carried out a pilot screen that identified three drugs (ibutilide, azaperone, and azelastine) as drugs that increase KV11.1 trafficking. Azaperone 115-124 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-171 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. VU0405601 158-167 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-138 35125346-8 2022 To validate the optimized trafficking assay, we carried out a pilot screen that identified three drugs (ibutilide, azaperone, and azelastine) as drugs that increase KV11.1 trafficking. azelastine 130-140 potassium voltage-gated channel subfamily H member 2 Homo sapiens 165-171 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. Cesium 176-182 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-97 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. Cesium 176-182 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-138 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. Cesium 184-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-97 35125346-7 2022 Results: The combination of: 1) truncating the trafficking-deficient variant KV11.1-G601S (KV11.1-G601S-G965*X), and addition of 2) KV11.1 channel activator (VU0405601) and 3) cesium (Cs+) to the Tl+ flux assay buffer resulted in an outstanding Z" of 0.83. Cesium 184-187 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-138 35087712-6 2022 The pro-arrhythmic action results from drug-induced inhibition of Kv11.1 (hERG) channels interfering with the repolarizing potassium IKr currents, leading to long QT and increased risk of triggered arrhythmias. Potassium 123-132 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-72 34025432-8 2021 Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. Ajmaline 123-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 33945158-4 2022 Inhibition of Eag1 reduces tumor growth, but the search for potent inhibitors for tumor therapy suffers from the structural similarities with the cardiac HERG channel, a major off-target. eag1 14-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 154-158 34025432-8 2021 Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. Ivabradine 133-143 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 34025432-8 2021 Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. Quinidine 145-154 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 34025432-8 2021 Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. Mexiletine 159-169 potassium voltage-gated channel subfamily H member 2 Homo sapiens 173-177 33658490-2 2021 In this work, we combine molecular dynamics simulations, mutagenesis, and electrophysiology to provide mechanistic insights into how lipophilic molecules (ceramide-sphingolipid probe) alter gating kinetics and K+ currents of hERG1. Ceramides 155-163 potassium voltage-gated channel subfamily H member 2 Homo sapiens 225-230 33829343-1 2021 The human ether-a-go-go related gene (hERG, KCNH2) encodes the pore-forming subunit of the potassium channel responsible for a fast component of the cardiac delayed rectifier potassium current (IKr). Potassium 91-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-49 33854869-3 2021 In addition to these apparent assets of Bedaquiline, potential disadvantages of Bedaquiline include inhibition of the hERG (human Ether-a-go-related gene; KCNH2), potassium channel (concurrent risk of cardiac toxicity), and risk of phospholipidosis due to its more lipophilic nature. bedaquiline 80-91 potassium voltage-gated channel subfamily H member 2 Homo sapiens 155-160 33722764-7 2021 Moreover, a drug-induced LQT was modelled by treating the pigs with the hERG1 blocker dofetilide (DOF). dofetilide 86-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 33722764-7 2021 Moreover, a drug-induced LQT was modelled by treating the pigs with the hERG1 blocker dofetilide (DOF). dofetilide 98-101 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 33658490-2 2021 In this work, we combine molecular dynamics simulations, mutagenesis, and electrophysiology to provide mechanistic insights into how lipophilic molecules (ceramide-sphingolipid probe) alter gating kinetics and K+ currents of hERG1. Sphingolipids 164-176 potassium voltage-gated channel subfamily H member 2 Homo sapiens 225-230 33658490-3 2021 We show that the sphingolipid probe induced a significant left shift of activation voltage, faster deactivation rates, and current blockade comparable to traditional hERG1 blockers. Sphingolipids 17-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 166-171 33611903-18 2021 Mexiletine significantly improved symptoms in 2 patients with LQT2 post ineffective beta-blocker medication. Mexiletine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 33611903-17 2021 Nadolol succeeded in eliminating cardiac events in one patient with LQT2 post ineffective metoprolol medication. Nadolol 0-7 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 33040444-0 2021 Letrozole targets the Human ether-a-go-go-related gene (HERG) potassium current in glioblastoma. Letrozole 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 33040444-0 2021 Letrozole targets the Human ether-a-go-go-related gene (HERG) potassium current in glioblastoma. Potassium 62-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 33261899-2 2021 We synthesized and evaluated three novel series of substituted benzophenones for their allosteric modulation of the human Kv11.1 (hERG) channel. Benzophenones 63-76 potassium voltage-gated channel subfamily H member 2 Homo sapiens 122-128 33381033-5 2020 In conclusion, nadolol was recommended as a relatively effective strategy for LQT2 in order to improve the prognosis of patients during a long follow-up period. Nadolol 15-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 78-82 33196188-10 2020 The original SILCS model was based on the all-atom modeling of the hERG1 channel in an explicit lipid bilayer and was further augmented with a Bayesian-optimization/machine-learning (BML) stage employing an independent literature-derived training set of 163 molecules. Lipid Bilayers 96-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 67-72 33452554-0 2021 Influence of Kv11.1 (hERG1) K+ channel expression on DNA damage induced by the genotoxic agent methyl methanesulfonate. Methyl Methanesulfonate 95-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-19 33452554-0 2021 Influence of Kv11.1 (hERG1) K+ channel expression on DNA damage induced by the genotoxic agent methyl methanesulfonate. Methyl Methanesulfonate 95-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-26 33452554-6 2021 Moreover, direct DNA damage measurements, using the comet assay, demonstrated for the first time that Kv11.1 conduction activity was able to modify MMS-induced DNA damage, decreasing it particularly at high MMS concentration, in a way related to PARP1 gene expression. Methyl Methanesulfonate 148-151 potassium voltage-gated channel subfamily H member 2 Homo sapiens 102-108 33467093-5 2021 In this study, we tested the hypothesis that the RNA polyadenylate binding protein nuclear 1 (PABPN1) interacts with a unique 22 nt adenosine stretch adjacent to the intron 9 poly(A) signal and regulates KCNH2 pre-mRNA alternative polyadenylation and the relative expression of Kv11.1a C-terminal isoforms. Adenosine 132-141 potassium voltage-gated channel subfamily H member 2 Homo sapiens 204-209 32587517-8 2020 Blocking KV11.1 with E4031 shows that channel activity contributed to modulate the beta1 integrin-dependent pHi increase. E 4031 21-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-15 32253972-2 2020 A frequent cause for LQTS are mutations in the KCNH2 gene (also known as the human ether-a-go-go-related gene or hERG), which reduce or modulate the potassium current IKr and hence alter cardiac repolarization. ether-a-go 83-93 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-52 32253972-2 2020 A frequent cause for LQTS are mutations in the KCNH2 gene (also known as the human ether-a-go-go-related gene or hERG), which reduce or modulate the potassium current IKr and hence alter cardiac repolarization. Potassium 149-158 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-52 33154722-3 2020 The aim of this study was to investigate the mechanisms of disopyramide underlying its antiarrhythmic effects in SQTS1 with the N588K mutation in HERG channel. Disopyramide 59-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 146-150 33154722-10 2020 The results demonstrated that disopyramide may be effective for preventing tachyarrhythmias in SQTS1-patients carrying the N588K mutation in HERG channel by APD-prolongation via enhancing ICa-L, late INa, INCX, and reducing ISK. Disopyramide 30-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 141-145 33002116-6 2021 METHODS AND RESULTS: Meta-analysis of 3653 public human RNA-seq datasets identified a strong correlation between expression of CACNA1C (L-type calcium current, ICaL) and KCNH2 (rapid delayed rectifier K+ current, IKr), which was also observed in human adult heart tissue samples. Calcium 143-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 170-175 32940533-3 2020 Recently, FDA-approved cystic fibrosis protein trafficking chaperone, lumacaftor (LUM), has been proposed as novel therapy for LQT2. lumacaftor 70-80 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 32940533-3 2020 Recently, FDA-approved cystic fibrosis protein trafficking chaperone, lumacaftor (LUM), has been proposed as novel therapy for LQT2. lumacaftor 82-85 potassium voltage-gated channel subfamily H member 2 Homo sapiens 127-131 32940533-4 2020 Here, we test the efficacy of LUM treatment in patient-specific induced pluripotent stem cell-cardiomyocytes (iPSC-CMs) derived from two patients with known LQT2 trafficking defective mutations and a patient with novel KCNH2 variant, p.R685P. lumacaftor 30-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 157-161 32940533-4 2020 Here, we test the efficacy of LUM treatment in patient-specific induced pluripotent stem cell-cardiomyocytes (iPSC-CMs) derived from two patients with known LQT2 trafficking defective mutations and a patient with novel KCNH2 variant, p.R685P. lumacaftor 30-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 219-224 32940533-10 2020 While LUM treatment rescued the phenotype of KCNH2-N633S and KCNH2-R685P, LUM paradoxically prolonged the APD90 in KCNH2-G604S iPSC-CMs. lumacaftor 6-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-50 32940533-10 2020 While LUM treatment rescued the phenotype of KCNH2-N633S and KCNH2-R685P, LUM paradoxically prolonged the APD90 in KCNH2-G604S iPSC-CMs. lumacaftor 6-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-66 32940533-10 2020 While LUM treatment rescued the phenotype of KCNH2-N633S and KCNH2-R685P, LUM paradoxically prolonged the APD90 in KCNH2-G604S iPSC-CMs. lumacaftor 6-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-66 32710951-5 2020 Sevoflurane, but not propofol, prolonged ventricular action potential duration and QT interval in wild-type, LQT1 and LQT2 models. Sevoflurane 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 118-122 32922187-11 2020 Molecular docking analysis successfully combined KCNH2, NCOA1, KDR and ADRB2 to Myricanone with docking scores from 4.61 to 6.28. myricanone 80-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 49-54 32694995-7 2020 Using multi-microseconds Molecular Dynamics (MD) simulations of wild-type and M651T mutant hERG1, we suggested the block of the channel through the lipid mediated pathway, the opening of which is facilitated by the flexible phenylalanine ring (F656). Phenylalanine 224-237 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-96 32534368-7 2020 To achieve therapeutic targeting, DTX-Au-PEG complex and DTX IN PEG-AuNPs were chemical combined with the human anti-EGFR polyclonal antibody, which recognizes the hERG1 channel aberrantly expressed on the membrane of human lung cancer cells. Docetaxel 34-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 164-169 32710951-6 2020 The QT-prolonging effect of sevoflurane was more profound in LQT2 than in wild-type and LQT1 models. Sevoflurane 28-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-65 32710951-8 2020 In LQT2 model, IKs was considerably enhanced during excessive prolongation of ventricular action potential duration by reduction of IKr and relative contribution of IKs to ventricular repolarization was markedly elevated, which appears to underlie more pronounced QT-prolonging effect of sevoflurane in LQT2 model, compared with wild-type and LQT1 models. Sevoflurane 288-299 potassium voltage-gated channel subfamily H member 2 Homo sapiens 3-7 32710951-9 2020 This simulation study clearly elucidates the electrophysiological basis underlying the difference in QT-prolonging effect of sevoflurane among wild-type, LQT1 and LQT2 models, and may provide important information for developing anesthetic strategies for patients with long QT syndrome in clinical settings. Sevoflurane 125-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 163-167 32533968-2 2020 In this study, Procyanidin B1, a natural compound extracted from the grape seed, was identified as a potent, specific inhibitor, which can inhibit the Kv10.1 channel in a concentration-dependent manner (IC50 = 10.38 +- 0.87 muM), but has negligible effects on other potassium channels, including Kir2.1, HERG or KCNQ1. procyanidin B1 15-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 304-308 33304417-6 2020 This is the first report that demonstrates the efficacy and safety of trans-maternal administration of nadolol for treatment of symptomatic LQT2 fetuses with TdP. Nadolol 103-110 potassium voltage-gated channel subfamily H member 2 Homo sapiens 140-144 32694995-9 2020 We investigated two well-established hERG1 blockers (ivabradine and dofetilide) for M651 sensitivity through electrophysiology and mutagenesis techniques. Ivabradine 53-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-42 32694995-9 2020 We investigated two well-established hERG1 blockers (ivabradine and dofetilide) for M651 sensitivity through electrophysiology and mutagenesis techniques. dofetilide 68-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-42 32694995-11 2020 Moreover, we show that the dofetilide-induced block of hERG1 occurs through the intracellular space, whereas little to no block of ivabradine is observed during the intracellular application of the drug. dofetilide 27-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 55-60 32231201-0 2020 Correction: Clarithromycin inhibits autophagy in colorectal cancer by regulating the hERG1 potassium channel interaction with PI3K. Clarithromycin 12-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 85-90 32173522-3 2020 Here, HEK293 cells were transfected with the HERG gene alone or co-transfected with HERG and AKAP5 using Lipofectamine 2000. Lipofectamine 105-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 84-88 32390841-6 2020 Attenuating the K+ channel function by applying the hERG1 channel inhibitor E4031 modulated Ca2+ signaling, impaired inhibition of the mitosis promoting subunit cdc2, overrode cell cycle arrest, and decreased clonogenic survival of the irradiated cells but did not affect repair of DNA double strand breaks suggesting a critical role of the hERG1 K+ channels for the Ca2+ signaling and the cell cycle control during DNA damage response. E 4031 76-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 52-57 32390841-6 2020 Attenuating the K+ channel function by applying the hERG1 channel inhibitor E4031 modulated Ca2+ signaling, impaired inhibition of the mitosis promoting subunit cdc2, overrode cell cycle arrest, and decreased clonogenic survival of the irradiated cells but did not affect repair of DNA double strand breaks suggesting a critical role of the hERG1 K+ channels for the Ca2+ signaling and the cell cycle control during DNA damage response. E 4031 76-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 341-346 32191791-0 2020 Structure of KCNH2 cyclic nucleotide-binding homology domain reveals a functionally vital salt-bridge. Nucleotides, Cyclic 19-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-18 32191791-2 2020 Like other KCNH family members, hKCNH2 channels contain a unique intracellular complex, consisting of an N-terminal eag domain and a C-terminal cyclic nucleotide-binding homology domain (CNBHD), which is crucial for channel function. Methyl 2-acetamido-2-deoxy-beta-D-glucopyranoside 116-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-38 32191791-2 2020 Like other KCNH family members, hKCNH2 channels contain a unique intracellular complex, consisting of an N-terminal eag domain and a C-terminal cyclic nucleotide-binding homology domain (CNBHD), which is crucial for channel function. Nucleotides, Cyclic 144-161 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-38 31901677-0 2020 Elucidation of interaction mechanism of hERG1 potassium channel with scorpion toxins BeKm-1 and BmTx3b. Potassium 46-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-45 31877041-5 2020 ICA-105574 (ICA), a 3-Nitro-n-(4-phenoxyphenyl) benzamide derivative activates hERG1 by strongly attenuating pore-type inactivation. 3-nitro-N-(4-phenoxyphenyl)benzamide 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-84 32391435-2 2020 Our findings indicate that the antibiotic clarithromycin can target hERG1 and modulate autophagy to promote the death of chemoresistant colorectal cancer cells. Clarithromycin 42-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-73 32123164-0 2020 Clarithromycin inhibits autophagy in colorectal cancer by regulating the hERG1 potassium channel interaction with PI3K. Clarithromycin 0-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-78 32123164-4 2020 Because Cla is known to bind human Ether-a-go-go Related Gene 1 (hERG1) K+ channels, we studied if its effects depended on hERG1 and its conformational states. Clarithromycin 8-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-70 32123164-4 2020 Because Cla is known to bind human Ether-a-go-go Related Gene 1 (hERG1) K+ channels, we studied if its effects depended on hERG1 and its conformational states. Clarithromycin 8-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 123-128 32123164-5 2020 By availing of hERG1 mutants with different gating properties, we found that fluorescently labelled Cla preferentially bound to the closed channels. Clarithromycin 100-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 15-20 32123164-6 2020 Furthermore, by sequestering the channel in the closed conformation, Cla inhibited the formation of a macromolecular complex between hERG1 and the p85 subunit of PI3K. Clarithromycin 69-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-138 32123164-9 2020 We conclude that Cla affects the autophagic flux by impairing the signaling pathway linking hERG1 and PI3K. Clarithromycin 17-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 92-97 31472168-10 2020 The hERG1 (KV11.1) blocker, E4031, followed by the IKs blocker, JNJ303, increased extracellular field potential duration in CPC-iPSC-CMs to a greater extent than in BMC- and HDF-iPSC-CMs. bmc 165-168 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-9 31557540-1 2020 BACKGROUND: KCNH2 encodes the human ether-a-go-go-related gene (hERG) potassium channel, which passes the rapid delayed rectifier potassium current, IKr. Potassium 70-79 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-17 31557540-1 2020 BACKGROUND: KCNH2 encodes the human ether-a-go-go-related gene (hERG) potassium channel, which passes the rapid delayed rectifier potassium current, IKr. Potassium 130-139 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-17 31877041-5 2020 ICA-105574 (ICA), a 3-Nitro-n-(4-phenoxyphenyl) benzamide derivative activates hERG1 by strongly attenuating pore-type inactivation. 3-nitro-N-(4-phenoxyphenyl)benzamide 20-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-84 32071644-0 2020 Mexiletine shortens the QT interval in a pedigree of KCNH2 related long QT syndrome. Mexiletine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 53-58 31948476-0 2020 The ERG1a potassium channel increases basal intracellular calcium concentration and calpain activity in skeletal muscle cells. Potassium 10-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 31948476-0 2020 The ERG1a potassium channel increases basal intracellular calcium concentration and calpain activity in skeletal muscle cells. Calcium 58-65 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 31570775-5 2020 Translating to humans, we find analogous dysfunctional interactions between hippocampus and prefrontal cortex in coupling of the fMRI blood oxygen level-dependent (BOLD) signal during working memory in healthy subjects carrying alleles associated with increased KCNH2-3.1 expression in brain. Oxygen 140-146 potassium voltage-gated channel subfamily H member 2 Homo sapiens 262-267 31751991-6 2020 This study focuses on investigating the potential variants on SCN5A, KCNQ1, and KCNH2 contributing to AMI with VA in a Chinese population. Vanillic Acid 111-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-85 30826123-8 2019 In this study we show that the activation kinetics in iPSC-CMs resemble hERG1b kinetics using Cs+ as a charge carrier. Cesium 94-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 31874991-3 2019 The human ether-a-go-go-related gene 1 (hERG1) encodes the pore-forming subunit underlying cardiac rapidly delayed rectifier potassium current (IKr). Potassium 125-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-45 31874991-7 2019 Quantitative RT-PCR, Western blot analysis, immunofluorescence and co-immunoprecipitation methods were used to determine the effects of mitragynine on hERG1a/1b expression and hERG1-cytosolic chaperones interaction. mitragynine 136-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 151-156 31664867-1 2019 There is significant interest in the potential utility of small molecule activator compounds to mitigate cardiac arrhythmia caused by loss-of-function of hERG1a voltage-gated potassium channels. Potassium 175-184 potassium voltage-gated channel subfamily H member 2 Homo sapiens 154-159 31664867-5 2019 Our functional data from isolated zkcnh6a channels show that these channels respond to hERG1a channel blockers (dofetilide and terfenadine), and type 1 (RPR260243) and type 2 (NS1643, PD-118057) hERG1a activator compounds, in a similar manner to hKCNH2a channels, with minor differences largely accounted for by subtly different biophysical properties in the two channels. dofetilide 112-122 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-92 31664867-5 2019 Our functional data from isolated zkcnh6a channels show that these channels respond to hERG1a channel blockers (dofetilide and terfenadine), and type 1 (RPR260243) and type 2 (NS1643, PD-118057) hERG1a activator compounds, in a similar manner to hKCNH2a channels, with minor differences largely accounted for by subtly different biophysical properties in the two channels. Terfenadine 127-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-92 31573043-5 2019 However, how the sequence context influences the efficiency of aminoglycosides in rescuing the human ether-a-go-go-related (HERG) protein in mammalian cells remains to be fully elucidated. Aminoglycosides 63-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 124-128 31573043-6 2019 Therefore, the present study was devised to examine the susceptibility of different termination codons on the HERG gene and the +4 nucleotide immediately following them to be suppressed by aminoglycosides in 293 cells. Aminoglycosides 189-204 potassium voltage-gated channel subfamily H member 2 Homo sapiens 110-114 31573043-8 2019 The read-through effect was subsequently examined by adding aminoglycoside G418 into the culture medium, followed by incubation of the cells for 24 h. An immunofluorescence method was then used to observe the protein expression of HERG prior to and following drug treatment. Aminoglycosides 60-74 potassium voltage-gated channel subfamily H member 2 Homo sapiens 231-235 31573043-13 2019 The level of red fluorescence was observed prior to and following the administration of G418 using antibodies targeting the N- or C-terminus of the HERG protein. Nitrogen 124-125 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 31573043-13 2019 The level of red fluorescence was observed prior to and following the administration of G418 using antibodies targeting the N- or C-terminus of the HERG protein. Carbon 130-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 148-152 31447465-0 2019 Mexiletine Suppressed Recurrent Ventricular Tachycardia Triggered by Hemodialysis in an Old Patient with LQT2. Mexiletine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-109 31807018-0 2019 The hERG1 Potassium Channel Behaves As Prognostic Factor In Gastric Dysplasia Endoscopic Samples. Potassium 10-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-9 30947366-6 2019 Ivabradine, ajmaline, and mexiletine inhibited KCNH2 channel currents significantly, which may underlie their APD-prolonging effects. Ivabradine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-52 31484079-1 2019 The human ether-a-go-go-related gene KCNH2 encodes the voltage-gated potassium channel underlying IKr, a current critical for the repolarization phase of the cardiac action potential. ether-a 10-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-42 30947366-6 2019 Ivabradine, ajmaline, and mexiletine inhibited KCNH2 channel currents significantly, which may underlie their APD-prolonging effects. Ajmaline 12-20 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-52 30947366-6 2019 Ivabradine, ajmaline, and mexiletine inhibited KCNH2 channel currents significantly, which may underlie their APD-prolonging effects. Mexiletine 26-36 potassium voltage-gated channel subfamily H member 2 Homo sapiens 47-52 31946488-2 2019 The SQT1, SQT2 and SQT3 variants of the SQTS result from inherited gain-of-function mutations (e.g. N588K, V307L and D172N, respectively) to potassium channels. Potassium 141-150 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-8 31182542-0 2019 The Pore-Lipid Interface: Role of Amino-Acid Determinants of Lipophilic Access by Ivabradine to the hERG1 Pore Domain. Ivabradine 82-92 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-105 31347270-0 2019 Long QT syndrome in chromosome 7q35q36.3 deletion involving KCNH2 gene: Warning for chlorpheniramine prescription. Chlorpheniramine 84-100 potassium voltage-gated channel subfamily H member 2 Homo sapiens 60-65 31347270-3 2019 RESULTS: Among the deleted genes, two genes have cardiac implications: PRKAG2 (OMIM #602743), associated with hypertrophic cardiomyopathy, cardiac conduction disease, and sudden death, and KCNH2 (OMIM #152427), coding for a cardiac potassium channel involved in long QT syndrome, unmasked by the chlorpheniramine treatment. Chlorpheniramine 296-312 potassium voltage-gated channel subfamily H member 2 Homo sapiens 189-194 31032878-5 2019 EXPERIMENTAL APPROACH: We analysed sensitivity of recombinant Kv 12.1 channels to quinine, a substituted quinoline that blocks Kv 10.1 and Kv 11.1 at low micromolar concentrations. Quinine 82-89 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-146 31032878-5 2019 EXPERIMENTAL APPROACH: We analysed sensitivity of recombinant Kv 12.1 channels to quinine, a substituted quinoline that blocks Kv 10.1 and Kv 11.1 at low micromolar concentrations. quinoline 105-114 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-146 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Quinine 50-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-197 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Quinine 50-57 potassium voltage-gated channel subfamily H member 2 Homo sapiens 208-215 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Histidine 98-107 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-197 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Histidine 98-107 potassium voltage-gated channel subfamily H member 2 Homo sapiens 208-215 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Tyrosine 178-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-197 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Tyrosine 178-186 potassium voltage-gated channel subfamily H member 2 Homo sapiens 208-215 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Quinine 221-228 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-197 31032878-8 2019 Low sensitivity of Kv 12.1 and characteristics of quinine-dependent inhibition were determined by histidine 462, as site-directed mutagenesis of this residue into the homologous tyrosine of Kv 11.1 conferred Kv 11.1-like quinine block to Kv 12.1(H462Y). Quinine 221-228 potassium voltage-gated channel subfamily H member 2 Homo sapiens 208-215 31032878-10 2019 In contrast, more favourable interactions can explain the higher quinine sensitivity of Kv 12.1(H462Y) and Kv 11.1 channels. Quinine 65-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 107-114 31946488-4 2019 In this study, computational modelling was used to investigate pharmacotherapeutic effects of selected class I drug quinidine on SQT1, SQT2 and SQT3 variants. Quinidine 116-125 potassium voltage-gated channel subfamily H member 2 Homo sapiens 129-133 31946488-10 2019 At the 10 muM concentration tested in this study, quinidine effectively prolonged the action potential duration (APD) under all the SQT1, SQT2 and SQT3 conditions. Quinidine 50-59 potassium voltage-gated channel subfamily H member 2 Homo sapiens 132-136 31275424-6 2019 The results of molecular docking showed that linalool, caryophyllene, dibutyl phthalate, (-)-4-terpineol, and (-)-alpha-terpineol have good binding activity with ADRB2, DRD2, ESR1, KCNH2, NR1H4, NR1I2, NR1I3, and TRPV1 targets. linalool 45-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-186 30938413-7 2019 Oxytocin prolonged QTc and steepened QT/RR-slope in vivo and prolonged ex vivo APD75 in LQT2 hearts. Oxytocin 0-8 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-92 31275424-6 2019 The results of molecular docking showed that linalool, caryophyllene, dibutyl phthalate, (-)-4-terpineol, and (-)-alpha-terpineol have good binding activity with ADRB2, DRD2, ESR1, KCNH2, NR1H4, NR1I2, NR1I3, and TRPV1 targets. alpha-terpineol 110-129 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-186 31275424-6 2019 The results of molecular docking showed that linalool, caryophyllene, dibutyl phthalate, (-)-4-terpineol, and (-)-alpha-terpineol have good binding activity with ADRB2, DRD2, ESR1, KCNH2, NR1H4, NR1I2, NR1I3, and TRPV1 targets. caryophyllene 55-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-186 31275424-6 2019 The results of molecular docking showed that linalool, caryophyllene, dibutyl phthalate, (-)-4-terpineol, and (-)-alpha-terpineol have good binding activity with ADRB2, DRD2, ESR1, KCNH2, NR1H4, NR1I2, NR1I3, and TRPV1 targets. Dibutyl Phthalate 70-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-186 31275424-6 2019 The results of molecular docking showed that linalool, caryophyllene, dibutyl phthalate, (-)-4-terpineol, and (-)-alpha-terpineol have good binding activity with ADRB2, DRD2, ESR1, KCNH2, NR1H4, NR1I2, NR1I3, and TRPV1 targets. (-)-Terpinen-4-ol 89-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 181-186 30689740-3 2019 The generally accepted common mechanism whereby drugs prolong QT is block of a key repolarizing potassium current in heart, IKr, generated by expression of KCNH2, also known as HERG. Potassium 96-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 156-161 31186788-0 2019 hERG1 is involved in the pathophysiological process and inhibited by berberine in SKOV3 cells. Berberine 69-78 potassium voltage-gated channel subfamily H member 2 Homo sapiens 0-5 31186788-11 2019 In addition, berberine (BBR) may be used as a potential drug in the treatment of ovarian cancer, possibly due to its inhibitory effects on the hERG1 channels. Berberine 13-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 143-148 31186788-11 2019 In addition, berberine (BBR) may be used as a potential drug in the treatment of ovarian cancer, possibly due to its inhibitory effects on the hERG1 channels. Berberine 24-27 potassium voltage-gated channel subfamily H member 2 Homo sapiens 143-148 31186788-12 2019 In conclusion, the present study demonstrated that hERG1 may be a potential therapeutic target in the treatment of ovarian cancer and provided novel insights into the mechanism underlying the antitumor effects of BBR in ovarian cancer. Berberine 213-216 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-56 31006312-12 2019 CONCLUSIONS: Although commonly prescribed in patients with LQT3, mexiletine also shortens the QTc significantly in two-thirds of a small subset of patients with potassium channel-mediated LQT2. Mexiletine 65-75 potassium voltage-gated channel subfamily H member 2 Homo sapiens 188-192 31006312-13 2019 In patients with LQT2, pharmacological targeting of the physiological late sodium current may provide added therapeutic efficacy to beta-blocker therapy. Sodium 75-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 30689740-3 2019 The generally accepted common mechanism whereby drugs prolong QT is block of a key repolarizing potassium current in heart, IKr, generated by expression of KCNH2, also known as HERG. Potassium 96-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 177-181 31169990-4 2019 Recent evidence, however, has shown significant affinity of ivabradine towards Kv11.1 (ether-a-go-go related gene, ERG) potassium channels. Ivabradine 60-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 79-85 30687112-8 2018 At the concentrations tested in this study, quinidine most effectively prolonged the APD and ERP in the setting of SQT1, followed by disopyramide and propafenone. Quinidine 44-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 115-119 30582453-10 2019 Moreover, QTc of SQT patient and action potential durations of SQT iPSC-CMs were both normalized by quinidine, indicating that quinidine is beneficial to KCNH2 T618I of SQT. Quinidine 100-109 potassium voltage-gated channel subfamily H member 2 Homo sapiens 154-159 30582453-10 2019 Moreover, QTc of SQT patient and action potential durations of SQT iPSC-CMs were both normalized by quinidine, indicating that quinidine is beneficial to KCNH2 T618I of SQT. Quinidine 127-136 potassium voltage-gated channel subfamily H member 2 Homo sapiens 154-159 31169990-5 2019 Despite the inhibition of Kv11.1 channels by ivabradine, cardiac action potential (AP) duration and heart rate corrected QT interval (QTc) of the human electrocardiogram (ECG) were not prolonged. Ivabradine 45-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 26-32 31169990-11 2019 Within this study we also confirm recent findings of human Kv11.1 inhibition by low microM concentrations of ivabradine and observed no prolongation of ventricular-like APs in cardiomyocytes derived from iPSCs. Ivabradine 109-119 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-65 31169990-12 2019 CONCLUSION: Our results provide an explanation why ivabradine, despite its affinity for Kv11.1 channels, does not prolong the cardiac AP and QTc interval. Ivabradine 51-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-94 29875689-6 2018 The microtubule depolymerizing drug nocodazole differentially affected G601S- and F805C-Kv11.1 protein immunostaining. Nocodazole 36-46 potassium voltage-gated channel subfamily H member 2 Homo sapiens 88-94 30567462-5 2019 RESULTS: Dofetilide and cisapride (IKr or Kv11.1 blockers) were associated with significant increases in QTca and JTpca, while sotalol was associated with significant increases in QTca, JTpca, and Tpeca. Cisapride 24-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-48 30567462-6 2019 Verapamil (a Kv11.1 and Cav1.2 blocker) resulted in a reduction in QTca and JTpca, however, and increased Tpeca. Verapamil 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 13-19 30405887-5 2018 The resulting scFv-hERG1-Cys showed much higher stability and protein yield, increased affinity and more advantageous binding kinetics, compared to the "native" anti-hERG1scFv. Cysteine 25-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 19-24 30405887-6 2018 The scFv-hERG1-Cys was hence chosen and characterized: it showed a good binding to the native hERG1 antigen expressed on cells, was stable in serum and displayed a fast pharmacokinetic profile once injected intravenously in nude mice. Cysteine 15-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 9-14 30405887-6 2018 The scFv-hERG1-Cys was hence chosen and characterized: it showed a good binding to the native hERG1 antigen expressed on cells, was stable in serum and displayed a fast pharmacokinetic profile once injected intravenously in nude mice. Cysteine 15-18 potassium voltage-gated channel subfamily H member 2 Homo sapiens 94-99 30405887-8 2018 Finally, the in vivo distribution of an Alexa Fluor 750 conjugated scFv-hERG1-Cys was evaluated both in healthy and tumor-bearing nude mice, showing a good tumor-to-organ ratio, ideal for visualizing hERG1-expressing tumor masses in vivo. Alexa Fluor 750 40-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 30405887-8 2018 Finally, the in vivo distribution of an Alexa Fluor 750 conjugated scFv-hERG1-Cys was evaluated both in healthy and tumor-bearing nude mice, showing a good tumor-to-organ ratio, ideal for visualizing hERG1-expressing tumor masses in vivo. Alexa Fluor 750 40-55 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-205 30405887-8 2018 Finally, the in vivo distribution of an Alexa Fluor 750 conjugated scFv-hERG1-Cys was evaluated both in healthy and tumor-bearing nude mice, showing a good tumor-to-organ ratio, ideal for visualizing hERG1-expressing tumor masses in vivo. Cysteine 78-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 72-77 30405887-8 2018 Finally, the in vivo distribution of an Alexa Fluor 750 conjugated scFv-hERG1-Cys was evaluated both in healthy and tumor-bearing nude mice, showing a good tumor-to-organ ratio, ideal for visualizing hERG1-expressing tumor masses in vivo. Cysteine 78-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 200-205 30405887-9 2018 In conclusion, the scFv-hERG1-Cys possesses features which make it a suitable tool for application in cancer molecular imaging. Cysteine 30-33 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-29 30110354-3 2018 In contrast, ostruthin inhibited other K+ channels, e.g. human ether-a-go-go-related gene (HERG1), inward-rectifier (Kir2.1), voltage-gated (Kv1.4), and two-pore domain (TASK-1) at higher concentrations, without affecting voltage-gated potassium channel (KCNQ1 and 3). ostruthin 13-22 potassium voltage-gated channel subfamily H member 2 Homo sapiens 91-96 29617054-10 2018 Telmisartan-mediated prolongation of AP was attenuated in KCNH2 siRNA-transfected HL-1 cells. Telmisartan 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 58-63 30341381-3 2018 Here we used the voltage-dependent dipicrylamine (DPA)-induced quench of fluorescent proteins (FPS) linked to different positions at the cytoplasmic domains of KCNH2 (hERG) to gain some insights about the coarse structure of these channel parts. dipicrylamine 35-48 potassium voltage-gated channel subfamily H member 2 Homo sapiens 160-165 30341381-3 2018 Here we used the voltage-dependent dipicrylamine (DPA)-induced quench of fluorescent proteins (FPS) linked to different positions at the cytoplasmic domains of KCNH2 (hERG) to gain some insights about the coarse structure of these channel parts. dipicrylamine 50-53 potassium voltage-gated channel subfamily H member 2 Homo sapiens 160-165 29875689-7 2018 Nocodazole caused G601S-Kv11.1 protein to distribute into peripheral reticular structures, and it increased the diffuse immunostaining of F805C-Kv11.1 protein around the transitional ER subcompartments. Nocodazole 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-30 29875689-7 2018 Nocodazole caused G601S-Kv11.1 protein to distribute into peripheral reticular structures, and it increased the diffuse immunostaining of F805C-Kv11.1 protein around the transitional ER subcompartments. Nocodazole 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 144-150 29875689-9 2018 Incubating cells in MG132 minimally impacted G601S-Kv11.1 immunostaining, but it dramatically increased the diffuse immunostaining of F805C-Kv11.1 protein in the transitional ER. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 20-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-57 29875689-9 2018 Incubating cells in MG132 minimally impacted G601S-Kv11.1 immunostaining, but it dramatically increased the diffuse immunostaining of F805C-Kv11.1 protein in the transitional ER. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 20-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 140-146 29650123-8 2018 Intergenotype comparison showed that the risk for patients with LQT2 and LQT3 increased by 130% and 157% at any QTc duration versus patients with LQT1. qtc 112-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 29725305-5 2018 To test our method, we focused on 16 LQT2 mutations in the hERG Per-Arnt-Sim (PAS) domain that were previously studied via a widely used biochemical approach that compares levels of 135-kDa immature and 155-kDa fully glycosylated hERG protein to infer surface expression. Aminosalicylic Acid 78-81 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 29146210-7 2018 We also examined pharmacological rescue of homozygous mutant Kv11.1 current in cells treated with E-4031 or dofetilide. E 4031 98-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-67 29146210-7 2018 We also examined pharmacological rescue of homozygous mutant Kv11.1 current in cells treated with E-4031 or dofetilide. dofetilide 108-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 61-67 29479853-2 2018 Here, silica, titanium, zinc, and magnesium oxide nanoparticles are screened against human hERG (Kv 11.1) voltage-gated potassium channels under a whole-cell voltage clamp. Silicon Dioxide 6-12 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-104 29479853-2 2018 Here, silica, titanium, zinc, and magnesium oxide nanoparticles are screened against human hERG (Kv 11.1) voltage-gated potassium channels under a whole-cell voltage clamp. Magnesium Oxide 34-49 potassium voltage-gated channel subfamily H member 2 Homo sapiens 97-104 29449809-4 2018 As a proof of principle, we evaluated extensively lipid membrane partitioning of d-sotalol, well-known blocker of a cardiac potassium channel Kv11.1 encoded by the hERG gene, with reported substantial proclivity for arrhythmogenesis. Dexsotalol 81-90 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-148 29548277-0 2018 Common variants in the hERG (KCNH2) voltage-gated potassium channel are associated with altered fasting and glucose-stimulated plasma incretin and glucagon responses. Glucose 108-115 potassium voltage-gated channel subfamily H member 2 Homo sapiens 29-34 29161243-0 2018 The combined activation of KCa3.1 and inhibition of Kv11.1/hERG1 currents contribute to overcome Cisplatin resistance in colorectal cancer cells. Cisplatin 97-106 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-64 29203485-4 2018 These quinolines showed high probabilities of Caco2 permeability and human intestinal absorption and low probabilities of mutagenicity and of hERG1 inhibition. Quinolines 6-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 142-147 28743763-0 2017 A key role for peroxynitrite-mediated inhibition of cardiac ERG (Kv11.1) K+ channels in carbon monoxide-induced proarrhythmic early afterdepolarizations. Peroxynitrous Acid 15-28 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-71 30036649-9 2018 One additional patient had variance of unknown significance (VUS) in KCNH2 and another one in ANK2. 3-[(3~{a}~{S},4~{S},6~{a}~{R})-4-carboxy-2,3,4,5,6,6~{a}-hexahydro-1~{H}-pyrrolo[2,3-c]pyrrol-3~{a}-yl]propyl-$l^{3}-oxidanyl-bis(oxidanyl)boranuide 61-64 potassium voltage-gated channel subfamily H member 2 Homo sapiens 69-74 29255512-12 2017 Conclusions: Atenolol is an effective treatment for genetically proven LQT1 and LQT2 children and adolescents, with good tolerability. Atenolol 13-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 28987271-7 2017 For Kv11.1 channels (hERG1) the extracellularly accessible histidines H578 and H587 are CORM-2 targets. Histidine 59-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 4-10 28987271-7 2017 For Kv11.1 channels (hERG1) the extracellularly accessible histidines H578 and H587 are CORM-2 targets. Histidine 59-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 21-26 28987271-8 2017 The strong CO-independent action of CORM-2 on Kv11.1 and Kv1.5 channels can be completely abolished when CORM-2 is applied in the presence of an excess of free histidine or human serum albumin; cysteine and methionine are further potential targets. Histidine 160-169 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-52 28987271-8 2017 The strong CO-independent action of CORM-2 on Kv11.1 and Kv1.5 channels can be completely abolished when CORM-2 is applied in the presence of an excess of free histidine or human serum albumin; cysteine and methionine are further potential targets. Cysteine 194-202 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-52 28987271-8 2017 The strong CO-independent action of CORM-2 on Kv11.1 and Kv1.5 channels can be completely abolished when CORM-2 is applied in the presence of an excess of free histidine or human serum albumin; cysteine and methionine are further potential targets. Methionine 207-217 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-52 29089383-5 2017 We report 1) that hERG1a dominant-negative subunits suppress hERG1b currents (and vice versa), 2) that disulfide bonds form between single cysteine residues experimentally introduced into an extracellular loop of hERG1a and hERG1b subunits and produce hERG1a-hERG1b dimers, and 3) that hERG1a and hERG1b subunits tagged with fluorescent proteins that are FRET pairs exhibit robust energy transfer at the plasma membrane. Disulfides 103-112 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-23 29089383-5 2017 We report 1) that hERG1a dominant-negative subunits suppress hERG1b currents (and vice versa), 2) that disulfide bonds form between single cysteine residues experimentally introduced into an extracellular loop of hERG1a and hERG1b subunits and produce hERG1a-hERG1b dimers, and 3) that hERG1a and hERG1b subunits tagged with fluorescent proteins that are FRET pairs exhibit robust energy transfer at the plasma membrane. Cysteine 139-147 potassium voltage-gated channel subfamily H member 2 Homo sapiens 18-23 28150511-8 2017 Since tadalafil blocks hERG1 K channels in concentration dependent manner, the cardiotoxicity prediction of the hit molecules was also tested. Tadalafil 6-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-28 28743763-9 2017 CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. onoo 64-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 196-202 28743763-9 2017 CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. onoo 64-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 247-253 28743763-0 2017 A key role for peroxynitrite-mediated inhibition of cardiac ERG (Kv11.1) K+ channels in carbon monoxide-induced proarrhythmic early afterdepolarizations. Carbon Monoxide 88-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-71 28743763-9 2017 CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. fetpps 81-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 196-202 28743763-9 2017 CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. fetpps 81-87 potassium voltage-gated channel subfamily H member 2 Homo sapiens 247-253 29059199-8 2017 Among the beta-blockers, nadolol showed a significant risk reduction in both LQT1 and LQT2 (HR 0.47 and 0.27, respectively), whereas atenolol and propranolol decreased the risk only in LQT1 (HR 0.36 and 0.46, respectively). Nadolol 25-32 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 28743763-9 2017 CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. 15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron 94-146 potassium voltage-gated channel subfamily H member 2 Homo sapiens 196-202 28768059-0 2017 (-)-Epicatechin rescues the As2 O3 -induced HERG K+ channel deficiency possibly through upregulating transcription factor SP1 expression. Catechin 0-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 28768059-0 2017 (-)-Epicatechin rescues the As2 O3 -induced HERG K+ channel deficiency possibly through upregulating transcription factor SP1 expression. Aligeron 28-31 potassium voltage-gated channel subfamily H member 2 Homo sapiens 44-48 28768059-5 2017 However, As2 O3 can induce the deficiency of HERG channel and cause LQT2. Arsenic Trioxide 9-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 68-72 28768059-8 2017 EPI was able to recover the protein expression and current of HERG channel disrupted by As2 O3 . Arsenic Trioxide 88-94 potassium voltage-gated channel subfamily H member 2 Homo sapiens 62-66 29059199-12 2017 Among them, nadolol was effective in LQT1 and LQT2, whereas other drugs showed different effectiveness depending on LQT genotype. Nadolol 12-19 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 28705808-0 2017 Molecular Basis of Altered hERG1 Channel Gating Induced by Ginsenoside Rg3. Ginsenosides 59-70 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-32 27581752-4 2017 Patch clamp experiments showed that the IC50 values of the human ether-a-go-go-related gene (hERG1) potassium (K) ion channel blocking affinity of PDE5 inhibitors sildenafil, vardenafil, and tadalafil as 33, 12, and 100 muM, respectively. Sildenafil Citrate 163-173 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-98 27581752-4 2017 Patch clamp experiments showed that the IC50 values of the human ether-a-go-go-related gene (hERG1) potassium (K) ion channel blocking affinity of PDE5 inhibitors sildenafil, vardenafil, and tadalafil as 33, 12, and 100 muM, respectively. Vardenafil Dihydrochloride 175-185 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-98 27581752-4 2017 Patch clamp experiments showed that the IC50 values of the human ether-a-go-go-related gene (hERG1) potassium (K) ion channel blocking affinity of PDE5 inhibitors sildenafil, vardenafil, and tadalafil as 33, 12, and 100 muM, respectively. Tadalafil 191-200 potassium voltage-gated channel subfamily H member 2 Homo sapiens 93-98 28705808-7 2017 Mutation to Ala of specific residues in the S1 (Tyr420), S2 (Leu452, Phe463), and S4 (Ile521, Lys525) segments partially inhibited the effects of Rg3 on hERG1. Alanine 12-15 potassium voltage-gated channel subfamily H member 2 Homo sapiens 153-158 28800628-1 2017 Many drugs used for non-cardiovascular and cardiovascular purposes, such as sotalol, have the side effect of prolonging cardiac repolarization, which can trigger life-threatening cardiac arrhythmias by inhibiting the potassium-channel IKr (KCNH2). Sotalol 76-83 potassium voltage-gated channel subfamily H member 2 Homo sapiens 240-245 29264083-5 2017 Paroxetine blocks the human ether-a-go-go-related gene (HERG) channels. Paroxetine 0-10 potassium voltage-gated channel subfamily H member 2 Homo sapiens 56-60 28963955-0 2017 Structural investigation of vesnarinone at the pore domains of open and open-inactivated states of hERG1 K+ channel. vesnarinone 28-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-104 28963955-1 2017 In this study, the dynamics of vesnarinone bounded hERG1 K+ channels are investigated using in silico approaches such as molecular docking, molecular dynamics (MD) simulations, MM/PBSA (Molecular Mechanics/Poisson Boltzmann Surface Area) calculations and Principal Component Analysis (PCA). vesnarinone 31-42 potassium voltage-gated channel subfamily H member 2 Homo sapiens 51-56 28593575-5 2017 Here, we investigated the role of ether-a-go-go-related 1 (hERG1) ion channels in the EMT of colorectal cancer cells. Ether 34-39 potassium voltage-gated channel subfamily H member 2 Homo sapiens 59-64 27466471-6 2016 In contrast, mutations in the cyclic nucleotide binding domain (cNBD) of KCNH2 conferred a negative risk of seizures, but not arrhythmias. Nucleotides, Cyclic 30-47 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-78 28500657-0 2017 Modulation of Kv 11.1 (hERG) channels by 5-(((1H-indazol-5-yl)oxy)methyl)-N-(4-(trifluoromethoxy)phenyl)pyrimidin-2-amine (ITP-2), a novel small molecule activator. 5-(((1h-indazol-5-yl)oxy)methyl)-n-(4-(trifluoromethoxy)phenyl)pyrimidin-2-amine 41-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-21 28500657-0 2017 Modulation of Kv 11.1 (hERG) channels by 5-(((1H-indazol-5-yl)oxy)methyl)-N-(4-(trifluoromethoxy)phenyl)pyrimidin-2-amine (ITP-2), a novel small molecule activator. itp-2 123-128 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-21 28632743-4 2017 Therefore, this study aimed to assess the potential effects of E-4031, disopyramide and quinidine on SQT1 using a mathematical model of human ventricular electrophysiology. E 4031 63-69 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 28632743-4 2017 Therefore, this study aimed to assess the potential effects of E-4031, disopyramide and quinidine on SQT1 using a mathematical model of human ventricular electrophysiology. Quinidine 88-97 potassium voltage-gated channel subfamily H member 2 Homo sapiens 101-105 28632743-15 2017 Quinidine exhibited significantly better therapeutic effects on SQT1 than E-4031 and disopyramide. Quinidine 0-9 potassium voltage-gated channel subfamily H member 2 Homo sapiens 64-68 28632743-16 2017 CONCLUSIONS: The simulated pharmacological actions of quinidine exhibited antiarrhythmic effects on SQT1. Quinidine 54-63 potassium voltage-gated channel subfamily H member 2 Homo sapiens 100-104 28632743-17 2017 This study substantiates a causal link between quinidine and QT interval prolongation in SQT1 and suggests that quinidine may be a potential pharmacological agent for treating SQT1 patients. Quinidine 47-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 28632743-17 2017 This study substantiates a causal link between quinidine and QT interval prolongation in SQT1 and suggests that quinidine may be a potential pharmacological agent for treating SQT1 patients. Quinidine 47-56 potassium voltage-gated channel subfamily H member 2 Homo sapiens 176-180 28632743-17 2017 This study substantiates a causal link between quinidine and QT interval prolongation in SQT1 and suggests that quinidine may be a potential pharmacological agent for treating SQT1 patients. Quinidine 112-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 89-93 28632743-17 2017 This study substantiates a causal link between quinidine and QT interval prolongation in SQT1 and suggests that quinidine may be a potential pharmacological agent for treating SQT1 patients. Quinidine 112-121 potassium voltage-gated channel subfamily H member 2 Homo sapiens 176-180 28525371-0 2017 Arsenic trioxide and angiotensin II have inhibitory effects on HERG protein expression: Evidence for the role of PML SUMOylation. Arsenic Trioxide 0-16 potassium voltage-gated channel subfamily H member 2 Homo sapiens 63-67 28525371-3 2017 Both arsenic trioxide (ATO) and angiotensin II (Ang II) were able to significantly reduce HERG protein expression, while also increasing PML SUMOylation and accelerating the formation of PML-NBs. Arsenic Trioxide 5-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 28525371-3 2017 Both arsenic trioxide (ATO) and angiotensin II (Ang II) were able to significantly reduce HERG protein expression, while also increasing PML SUMOylation and accelerating the formation of PML-NBs. Arsenic Trioxide 23-26 potassium voltage-gated channel subfamily H member 2 Homo sapiens 90-94 28525371-4 2017 Pre-exposure of cardiomyocytes to a SUMOylation chemical inhibitor, ginkgolic acid, or the silencing of UBC9 suppressed PML SUMOylation, subsequently preventing the downregulation of HERG induced by ATO or Ang II. ginkgolic acid 68-82 potassium voltage-gated channel subfamily H member 2 Homo sapiens 183-187 27394160-14 2016 CONCLUSIONS: Bisoprolol treatment results in QTc shortening in gene-positive LQT1 and LQT2 patients and is well tolerated during long-term administration. Bisoprolol 13-23 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 27761169-9 2016 The proteasome inhibitor MG132 increased the expression of immature A78T-HERG and increased both the immature and mature forms of WT-HERG. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 25-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-77 27761169-9 2016 The proteasome inhibitor MG132 increased the expression of immature A78T-HERG and increased both the immature and mature forms of WT-HERG. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 25-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 133-137 27761169-13 2016 Maturation of the A78T-HERG protein was facilitated by HS, expression of HSF-1, or exposure to geranyl geranyl acetone. geranylgeranylacetone 95-118 potassium voltage-gated channel subfamily H member 2 Homo sapiens 23-27 27517748-10 2016 Finally, a novel fluorophore- conjugated recombinant single chain variable fragment antibody (scFv-hERG1-Alexa488) was tested on freshly collected live BE biopsies: it could recognize hERG1 positive samples, perfectly matching IHC data.Overall, hERG1 can be considered a novel BE biomarker to be exploited for a novel endoscopic surveillance protocol, either in biopsies or through endoscopy, to identify those BE patients with higher risk to progress to EA. alexa488 105-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 184-189 27502018-1 2016 Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-a-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. Ginsenosides 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-110 27502018-1 2016 Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-a-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. Ginsenosides 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-118 27502018-1 2016 Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-a-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. steroid glycoside 33-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 105-110 27502018-1 2016 Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-a-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. steroid glycoside 33-50 potassium voltage-gated channel subfamily H member 2 Homo sapiens 112-118 27517748-10 2016 Finally, a novel fluorophore- conjugated recombinant single chain variable fragment antibody (scFv-hERG1-Alexa488) was tested on freshly collected live BE biopsies: it could recognize hERG1 positive samples, perfectly matching IHC data.Overall, hERG1 can be considered a novel BE biomarker to be exploited for a novel endoscopic surveillance protocol, either in biopsies or through endoscopy, to identify those BE patients with higher risk to progress to EA. alexa488 105-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 99-104 27517748-10 2016 Finally, a novel fluorophore- conjugated recombinant single chain variable fragment antibody (scFv-hERG1-Alexa488) was tested on freshly collected live BE biopsies: it could recognize hERG1 positive samples, perfectly matching IHC data.Overall, hERG1 can be considered a novel BE biomarker to be exploited for a novel endoscopic surveillance protocol, either in biopsies or through endoscopy, to identify those BE patients with higher risk to progress to EA. alexa488 105-113 potassium voltage-gated channel subfamily H member 2 Homo sapiens 184-189 26887900-8 2016 Following LCSD, there was a 57 +- 35 ms decrease in QTc in LQT1 (P = .16) and 23 +- 21 ms decrease in QTc in LQT2 (P = .3). lcsd 10-14 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 25945833-4 2016 We have demonstrated that the Kv11.1 channel activator NS1643 activates a calcineurin-dependent transcription of p21waf/cip and that this event is fundamental for the inhibitory effect of NS1643 on cell proliferation. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 55-61 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-36 25945833-4 2016 We have demonstrated that the Kv11.1 channel activator NS1643 activates a calcineurin-dependent transcription of p21waf/cip and that this event is fundamental for the inhibitory effect of NS1643 on cell proliferation. 1,3-bis(2-hydroxy-5-trifluoromethylphenyl)urea 188-194 potassium voltage-gated channel subfamily H member 2 Homo sapiens 30-36 26887900-8 2016 Following LCSD, there was a 57 +- 35 ms decrease in QTc in LQT1 (P = .16) and 23 +- 21 ms decrease in QTc in LQT2 (P = .3). qtc 102-105 potassium voltage-gated channel subfamily H member 2 Homo sapiens 109-113 27190211-7 2016 Here we present the first complete study of the temperature-dependent kinetics of block and unblock of a proarrhythmic drug, cisapride, to KV11.1. Cisapride 125-134 potassium voltage-gated channel subfamily H member 2 Homo sapiens 139-145 27555138-2 2016 We describe a case of torsade de pointes (TdP) caused by sevoflurane in a patient with c-LQTS genotype 2 (LQT2). Sevoflurane 57-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-104 27555138-2 2016 We describe a case of torsade de pointes (TdP) caused by sevoflurane in a patient with c-LQTS genotype 2 (LQT2). Sevoflurane 57-68 potassium voltage-gated channel subfamily H member 2 Homo sapiens 106-110 27555138-6 2016 Analysis of the electrocardiogram revealed that the corrected QT interval before anesthesia was 530 ms and 2.0% sevoflurane markedly prolonged the corrected QT interval to 693 ms. Postoperative studies revealed a mutation in the KCNH2 gene. Sevoflurane 112-123 potassium voltage-gated channel subfamily H member 2 Homo sapiens 229-234 26022375-7 2016 Later, genetic analysis demonstrated that this patient had a mutation in KCNH2 gene, and she was diagnosed as a type-2 long-QT syndrome which was accentuated by use of garenoxacin. garenoxacin 168-179 potassium voltage-gated channel subfamily H member 2 Homo sapiens 73-78