Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Toxicol Sci ; 154(1): 174-182, 2016 11.
Article in English | MEDLINE | ID: mdl-27503387

ABSTRACT

Drug-drug interactions pose a difficult drug safety problem, given the increasing number of individuals taking multiple medications and the relative complexity of assessing the potential for interactions. For example, sofosbuvir-based drug treatments have significantly advanced care for hepatitis C virus-infected patients, yet recent reports suggest interactions with amiodarone may cause severe symptomatic bradycardia and thus limit an otherwise extremely effective treatment. Here, we evaluated the ability of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) to recapitulate the interaction between sofosbuvir and amiodarone in vitro, and more generally assessed the feasibility of hiPSC-CMs as a model system for drug-drug interactions. Sofosbuvir alone had negligible effects on cardiomyocyte electrophysiology, whereas the sofosbuvir-amiodarone combination produced dose-dependent effects beyond that of amiodarone alone. By comparison, GS-331007, the primary circulating metabolite of sofosbuvir, had no effect alone or in combination with amiodarone. Further mechanistic studies revealed that the sofosbuvir-amiodarone combination disrupted intracellular calcium (Ca2+) handling and cellular electrophysiology at pharmacologically relevant concentrations, and mechanical activity at supra-pharmacological (30x Cmax) concentrations. These effects were independent of the common mechanisms of direct ion channel block and P-glycoprotein activity. These results support hiPSC-CMs as a comprehensive, yet scalable model system for the identification and evaluation of cardioactive pharmacodynamic drug-drug interactions.


Subject(s)
Amiodarone/toxicity , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Sofosbuvir/toxicity , Drug Interactions , Humans
2.
Tissue Eng Part C Methods ; 21(8): 852-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25654582

ABSTRACT

Cardiomyocytes (CMs) are terminally differentiated cells in the adult heart, and ischemia and cardiotoxic compounds can lead to cell death and irreversible decline of cardiac function. As testing platforms, isolated organs and primary cells from rodents have been the standard in research and toxicology, but there is a need for better models that more faithfully recapitulate native human biology. Hence, a new in vitro model comprising the advantages of 3D cell culture and the availability of induced pluripotent stem cells (iPSCs) of human origin was developed and characterized. Human CMs derived from iPSCs were studied in standard 2D culture and as cardiac microtissues (MTs) formed in hanging drops. Two-dimensional cultures were examined using immunofluorescence microscopy and western blotting, while the cardiac MTs were subjected to immunofluorescence, contractility, and pharmacological investigations. iPSC-derived CMs in 2D culture showed well-formed myofibrils, cell-cell contacts positive for connexin-43, and other typical cardiac proteins. The cells reacted to prohypertrophic growth factors with a substantial increase in myofibrils and sarcomeric proteins. In hanging drop cultures, iPSC-derived CMs formed spheroidal MTs within 4 days, showing a homogeneous tissue structure with well-developed myofibrils extending throughout the whole spheroid without a necrotic core. MTs showed spontaneous contractions for more than 4 weeks that were recorded by optical motion tracking, sensitive to temperature and responsive to electrical pacing. Contractile pharmacology was tested with several agents known to modulate cardiac rate and viability. Calcium transients underlay the contractile activity and were also responsive to electrical stimulation, caffeine-induced Ca(2+) release, and extracellular calcium levels. A three-dimensional culture using iPSC-derived human CMs provides an organoid human-based cellular platform that is free of necrosis and recapitulates vital cardiac functionality, thereby providing a new and promising relevant model for the evaluation and development of new therapies and detection of cardiotoxicity.


Subject(s)
Calcium Signaling , Induced Pluripotent Stem Cells/metabolism , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Spheroids, Cellular/metabolism , Tissue Scaffolds/chemistry , Adult , Connexin 43/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Spheroids, Cellular/cytology
3.
Am J Physiol Heart Circ Physiol ; 301(5): H2006-17, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21890694

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) can differentiate into functional cardiomyocytes; however, the electrophysiological properties of hiPSC-derived cardiomyocytes have yet to be fully characterized. We performed detailed electrophysiological characterization of highly pure hiPSC-derived cardiomyocytes. Action potentials (APs) were recorded from spontaneously beating cardiomyocytes using a perforated patch method and had atrial-, nodal-, and ventricular-like properties. Ventricular-like APs were more common and had maximum diastolic potentials close to those of human cardiac myocytes, AP durations were within the range of the normal human electrocardiographic QT interval, and APs showed expected sensitivity to multiple drugs (tetrodotoxin, nifedipine, and E4031). Early afterdepolarizations (EADs) were induced with E4031 and were bradycardia dependent, and EAD peak voltage varied inversely with the EAD take-off potential. Gating properties of seven ionic currents were studied including sodium (I(Na)), L-type calcium (I(Ca)), hyperpolarization-activated pacemaker (I(f)), transient outward potassium (I(to)), inward rectifier potassium (I(K1)), and the rapidly and slowly activating components of delayed rectifier potassium (I(Kr) and I(Ks), respectively) current. The high purity and large cell numbers also enabled automated patch-clamp analysis. We conclude that these hiPSC-derived cardiomyocytes have ionic currents and channel gating properties underlying their APs and EADs that are quantitatively similar to those reported for human cardiac myocytes. These hiPSC-derived cardiomyocytes have the added advantage that they can be used in high-throughput assays, and they have the potential to impact multiple areas of cardiovascular research and therapeutic applications.


Subject(s)
Cell Differentiation , Excitation Contraction Coupling , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Cell Line , Excitation Contraction Coupling/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Heart Rate , Humans , Induced Pluripotent Stem Cells/drug effects , Ion Channel Gating , Ion Transport , Kinetics , Membrane Transport Modulators/pharmacology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/metabolism , Sodium/metabolism , Sodium Channels/metabolism
4.
J Cardiovasc Transl Res ; 4(1): 66-72, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21061105

ABSTRACT

Moving from the bench to the bedside is an expensive and arduous journey with a high risk of failure. One roadblock on the path of translational medicine is the paucity of predictive in vitro models available during preclinical drug development. The ability of human embryonic stem (ES) and induced pluripotent stem (iPS) cells to generate virtually any tissue of the body, in vitro, makes these cells an obvious choice for use in drug discovery and translational medicine. Technological advancements in the production of stem cells and their differentiation into relevant cell types, such as cardiomyocytes, has permitted the utility of these cells in the translational medicine setting. In particular, the derivation and differentiation of patient-specific iPS cells will facilitate an understanding of basic disease etiology, enable better drug efficacy and safety screens, and ultimately lead to personalized patient therapies. This review will focus on recent advancements in the derivation and differentiation of human ES and iPS cells into cardiomyocytes and their uses in safety testing and modeling human disease.


Subject(s)
Cardiovascular Diseases/surgery , Embryonic Stem Cells/transplantation , Induced Pluripotent Stem Cells/transplantation , Myocytes, Cardiac/transplantation , Stem Cell Transplantation , Translational Research, Biomedical , Animals , Cardiovascular Diseases/pathology , Cell Differentiation , Cell Proliferation , Humans , Stem Cell Transplantation/adverse effects , Treatment Outcome
5.
Am J Physiol Heart Circ Physiol ; 298(6): H1842-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20363883

ABSTRACT

Mutations in human ether-a-go-go-related gene 1 (hERG) are linked to long QT syndrome type 2 (LQT2). hERG encodes the pore-forming alpha-subunits that coassemble to form rapidly activating delayed rectifier K(+) current in the heart. LQT2-linked missense mutations have been extensively studied in noncardiac heterologous expression systems, where biogenic (protein trafficking) and biophysical (gating and permeation) abnormalities have been postulated to underlie the loss-of-function phenotype associated with LQT2 channels. Little is known about the properties of LQT2-linked hERG channel proteins in native cardiomyocyte systems. In this study, we expressed wild-type (WT) hERG and three LQT2-linked mutations in neonatal mouse cardiomyocytes and studied their electrophysiological and biochemical properties. Compared with WT hERG channels, the LQT2 missense mutations G601S and N470D hERG exhibited altered protein trafficking and underwent pharmacological correction, and N470D hERG channels gated at more negative voltages. The DeltaY475 hERG deletion mutation trafficked similar to WT hERG channels, gated at more negative voltages, and had rapid deactivation kinetics, and these properties were confirmed in both neonatal mouse cardiomyocyte and human embryonic kidney (HEK)-293 cell expression systems. Differences between the cardiomyocytes and HEK-293 cell expression systems were that hERG current densities were reduced 10-fold and deactivation kinetics were accelerated 1.5- to 2-fold in neonatal mouse cardiomyocytes. An important finding of this work is that pharmacological correction of trafficking-deficient LQT2 mutations, as a potential innovative approach to therapy, is possible in native cardiac tissue.


Subject(s)
Animals, Newborn/metabolism , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Line , ERG1 Potassium Channel , Electrophysiological Phenomena , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Mice , Models, Animal , Mutation, Missense/genetics , Myocytes, Cardiac/cytology , Patch-Clamp Techniques
6.
Circulation ; 113(3): 365-73, 2006 Jan 24.
Article in English | MEDLINE | ID: mdl-16432067

ABSTRACT

BACKGROUND: The KCNH2 or human ether-a-go-go related gene (hERG) encodes the Kv11.1 alpha-subunit of the rapidly activating delayed rectifier K+ current (IKr) in the heart. Type 2 congenital long-QT syndrome (LQT2) results from KCNH2 mutations that cause loss of Kv11.1 channel function. Several mechanisms have been identified, including disruption of Kv11.1 channel synthesis (class 1), protein trafficking (class 2), gating (class 3), or permeation (class 4). For a few class 2 LQT2-Kv11.1 channels, it is possible to increase surface membrane expression of Kv11.1 current (IKv11.1). We tested the hypotheses that (1) most LQT2 missense mutations generate trafficking-deficient Kv11.1 channels, and (2) their trafficking-deficient phenotype can be corrected. METHODS AND RESULTS: Wild-type (WT)-Kv11.1 channels and 34 missense LQT2-Kv11.1 channels were expressed in HEK293 cells. With Western blot analyses, 28 LQT2-Kv11.1 channels had a trafficking-deficient (class 2) phenotype. For the majority of these mutations, the class 2 phenotype could be corrected when cells were incubated for 24 hours at reduced temperature (27 degrees C) or in the drugs E4031 or thapsigargin. Four of the 6 LQT2-Kv11.1 channels that had a wild-type-like trafficking phenotype did not cause loss of Kv11.1 function, which suggests that these channels are uncommon sequence variants. CONCLUSIONS: This is the first study to identify a dominant mechanism, class 2, for the loss of Kv11.1 channel function in LQT2 and to report that the class 2 phenotype for many of these mutant channels can be corrected. This suggests that if therapeutic strategies to correct protein trafficking abnormalities can be developed, it may offer clinical benefits for LQT2 patients.


Subject(s)
Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Long QT Syndrome/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Protein Transport/physiology , Cell Line , ERG1 Potassium Channel , Enzyme Inhibitors/pharmacology , Genes, Dominant , Humans , Kidney/cytology , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Mutation, Missense , Patch-Clamp Techniques , Phenotype , Protein Transport/drug effects , Thapsigargin/pharmacology
7.
Br J Pharmacol ; 147(6): 642-52, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16314852

ABSTRACT

beta-Blockers are widely used in the treatment of cardiovascular diseases. However, their effects on HERG channels at comparable conditions remain to be defined. We investigated the direct acute effects of beta-blockers on HERG current and the molecular basis of drug binding to HERG channels with mutations of putative common binding site (Y652A and F656C). beta-Blockers were selected based on the receptor subtype. Wild-type, Y652A and F656C mutants of HERG channel were stably expressed in HEK293 cells, and the current was recorded by using whole-cell patch-clamp technique (23 degrees C). 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). The IC(50) value for carvedilol was a clinically relevant concentration. High metoprolol (beta(1)-selective) concentrations were required for blockade (IC(50) 145 microM), and atenolol (beta(1)-selective) did not inhibit the HERG current. Inhibition of HERG current by carvedilol, propranolol and ICI 118551 was partially but significantly attenuated in Y652A and F656C mutant channels. Affinities of metoprolol to Y652A and F656C mutant channels were not different compared with the wild-type. HERG current block by all beta-blockers was not frequency-dependent. Drug affinities to HERG channels were different in beta-blockers. Our results provide additional strategies for clinical usage of beta-blockers. Atenolol and metoprolol may be preferable for patients with type 1 and 2 long QT syndrome. Carvedilol has a class III antiarrhythmic effect, which may provide the rationale for a favourable clinical outcome compared with other beta-blockers as suggested in the recent COMET (Carvedilol Or Metoprolol European Trial) substudy.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Adrenergic beta-Antagonists/metabolism , Adrenergic beta-Antagonists/therapeutic use , Binding Sites , Carbazoles/metabolism , Carbazoles/pharmacology , Carbazoles/therapeutic use , Cardiovascular Diseases/complications , Cardiovascular Diseases/drug therapy , Carvedilol , Cell Line , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Long QT Syndrome/complications , Long QT Syndrome/drug therapy , Membrane Potentials/drug effects , Metoprolol/metabolism , Metoprolol/pharmacology , Metoprolol/therapeutic use , Mutation , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/therapeutic use , Propanolamines/metabolism , Propanolamines/pharmacology , Propanolamines/therapeutic use , Propranolol/metabolism , Propranolol/pharmacology , Propranolol/therapeutic use , Protein Binding , Transfection
8.
Am J Physiol Heart Circ Physiol ; 290(3): H1278-88, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16227340

ABSTRACT

KCNH2 (hERG1) encodes the alpha-subunit proteins for the rapidly activating delayed rectifier K+ current (I(Kr)), a major K+ current for cardiac myocyte repolarization. In isolated myocytes I(Kr) frequently is small in amplitude or absent, yet KCNH2 channels and I(Kr) are targets for drug block or mutations to cause long QT syndrome. We hypothesized that KCNH2 channels and I(Kr) are uniquely sensitive to enzymatic damage. To test this hypothesis, we studied heterologously expressed K+, Na+, and L-type Ca2+ channels, and in ventricular myocytes I(Kr), slowly activating delayed rectifier K+ current (I(Ks)), and inward rectifier K+ current (I(K1)), by using electrophysiological and biochemical methods. 1) Specific exogenous serine proteases (protease XIV, XXIV, or proteinase K) selectively degraded KCNH2 current (I(KCNH2)) and its mature channel protein without damaging cell integrity and with minimal effects on the other channel currents; 2) immature KCNH2 channel protein remained intact; 3) smaller molecular mass KCNH2 degradation products appeared; 4) protease XXIV selectively abolished I(Kr); and 5) reculturing HEK-293 cells after protease exposure resulted in the gradual recovery of I(KCNH2) and its mature channel protein over several hours. Thus the channel protein for I(KCNH2) and I(Kr) is uniquely sensitive to proteolysis. Analysis of the degradation products suggests selective proteolysis within the S5-pore extracellular linker, which is structurally unique among Kv channels. These data provide 1) a new mechanism to account for low I(Kr) density in some isolated myocytes, 2) evidence that most complexly glycosylated KCNH2 channel protein is in the plasma membrane, and 3) new insight into the rate of biogenesis of KCNH2 channel protein within cells.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Ion Channel Gating/physiology , Membrane Potentials/physiology , Myocytes, Cardiac/metabolism , Potassium/metabolism , Serine Endopeptidases/administration & dosage , Animals , Cells, Cultured , Dogs , ERG1 Potassium Channel , Ion Channel Gating/drug effects , Membrane Potentials/drug effects
9.
Mol Pharmacol ; 68(1): 233-40, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15851652

ABSTRACT

Mutations in the KCNH2 or human ether-a-go-go-related gene-encoded K(+) channel reduce functional KCNH2 current (I(KCNH2)) to cause long QT syndrome (LQT2) by multiple mechanisms, including defects in intracellular transport (trafficking). Trafficking-deficient, or class 2, LQT2 mutations reduce the Golgi processing and surface membrane expression of KCNH2 channel proteins. Drugs that associate with pore-S6 intracellular drug binding domain of KCNH2 channel proteins to cause high-affinity block of I(KCNH2) also can increase the processing of class 2 LQT2 channel proteins through the secretory pathway. We used a strategy of intragenic suppression to test the hypothesis that amino acid substitutions in the putative drug binding domain at residue Y652 could compensate for protein folding abnormalities caused by class 2 LQT2 mutations. We found that the Y652C substitution, and to lesser extent the Y652S substitution, resulted in intragenic suppression of the class 2 LQT2 G601S phenotype; these substitutions increased Golgi processing of G601S channel proteins. The Y652C substitution also caused intragenic suppression of the class 2 LQT2 V612L and F640V phenotypes but not the LQT2 N470D or F805C phenotypes. These are the first findings to demonstrate that a single amino acid substitution in the putative KCNH2 drug binding domain can cause intragenic suppression of several LQT2 mutations.


Subject(s)
Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Suppression, Genetic , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Ion Channel Gating/genetics , Male , Middle Aged , Protein Transport/genetics
10.
Lancet ; 365(9460): 682-6, 2005.
Article in English | MEDLINE | ID: mdl-15721475

ABSTRACT

The HIV protease inhibitor class of antiretroviral drug causes unpredicted adverse effects by changing elements of normal cellular metabolism. A case of QT prolongation in a patient receiving protease inhibitors made us question whether these drugs might be responsible. We identified 24 patients with QT prolongation or torsade de pointes, or both, associated with protease inhibitors, using the Food and Drug Administration's voluntary adverse event reporting system. Attending physicians thought that protease inhibitors were the most probable cause of these symptoms in 14 of the patients. 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. We also recorded block by lopinavir of repolarising potassium current (I(Kr)) channels in neonatal mouse cardiac myocytes. Our data show that four protease inhibitors block HERG channels, suggesting that protease inhibitors could predispose individuals to QT prolongation and torsade de pointes.


Subject(s)
Cation Transport Proteins/metabolism , HIV Protease Inhibitors/adverse effects , Potassium Channels, Voltage-Gated/metabolism , Action Potentials , Animals , Cell Line , Electrocardiography , Ether-A-Go-Go Potassium Channels , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Heart/physiopathology , Humans , Kidney/metabolism , Long QT Syndrome/chemically induced , Long QT Syndrome/diagnosis , Male , Mice , Middle Aged , Myocardium/metabolism , Potassium Channel Blockers/adverse effects , Torsades de Pointes/chemically induced , Torsades de Pointes/diagnosis
11.
J Am Coll Cardiol ; 44(6): 1283-91, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15364333

ABSTRACT

OBJECTIVES: The purpose of this research was to determine whether an intronic variant (T1945+6C) in KCNH2 is a disease-causing mutation, and if expanded phenotyping criteria produce improved identification of long QT syndrome (LQTS) patients. BACKGROUND: Long QT syndrome is usually caused by mutations in conserved coding regions or invariant splice sites, yet no mutation is found in 30% to 50% of families. In one such family, we identified an intronic variant in KCNH2. Long QT syndrome diagnosis is hindered by reduced penetrance, as the long QT phenotype is absent on baseline electrocardiogram (ECG) in about 30%. METHODS: Fifty-two family members were phenotyped by baseline QTc, QTc maximum on serial ECGs (Ser QTc-max), and on exercise ECGs (Ex QTc-max) and by T-wave patterns. Linkage analysis tested association of the intronic change with phenotype. The consequences of T1945+6C on splicing was studied using a minigene system and on function by heterologous expression. RESULTS: Expanded phenotype/pedigree criteria identified 23 affected and 29 unaffected. Affected versus unaffected had baseline QTc 484 +/- 48 ms versus 422 +/- 20 ms, Ser QTc-max 508 +/- 48 ms versus 448 +/- 10 ms, Ex QTc-max 513 +/- 54 ms versus 444 +/- 11 ms, and LQT2 T waves in 87% versus 0%. Linkage analysis demonstrated a logarithm of odds score of 10.22. Splicing assay showed T1945+6C caused downstream intron retention. Complementary deoxyribonucleic acid with retained intron 7 failed to produce functional channels. CONCLUSIONS: T1945+6C is a disease-causing mutation. It alters KCNH2 splicing and cosegregates with the LQT2 phenotype. Expanded ECG criteria plus pedigree analysis provided accurate clinical diagnosis of all carriers including those with reduced penetrance. Intronic mutations may be responsible for LQTS in some families with otherwise negative mutation screening.


Subject(s)
Introns/genetics , Long QT Syndrome/genetics , Mutation/genetics , Potassium Channels, Voltage-Gated , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , ERG1 Potassium Channel , Electrocardiography , Ether-A-Go-Go Potassium Channels , Family Health , Follow-Up Studies , Genetic Carrier Screening , Genetic Linkage/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Pedigree , Phenotype , Potassium Channels/genetics , RNA, Complementary/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Statistics as Topic
12.
Circ Res ; 94(11): 1418-28, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15192037

ABSTRACT

The mechanisms underlying normal and abnormal cardiac rhythms are complex and incompletely understood. Through the study of uncommon inheritable arrhythmia syndromes, including the long QT and Brugada syndromes, new insights are emerging. At the cellular and tissue levels, we now recognize that ion channel current is the sum of biophysical (gating, permeation), biochemical (phosphorylation, etc), and biogenic (biosynthesis, processing, trafficking, and degradation) properties. This review focuses on how heart cells process ion channel proteins and how this protein trafficking may be altered in some cardiac arrhythmia diseases. In this review, we honor Dr Harry A. Fozzard, a modern pioneer in cardiac arrhythmias, cell biology, and molecular electrophysiology. As a scientist and physician, his writings and mentorship have served to foster a generation of investigators who continue to bring this complex field toward greater scientific understanding and impact on humankind.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Ion Channels/physiology , Protein Transport/physiology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , ERG1 Potassium Channel , Endoplasmic Reticulum/physiology , Ether-A-Go-Go Potassium Channels , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/physiopathology , Genetic Predisposition to Disease , Humans , Ion Channel Gating/physiology , Ion Channels/chemistry , Ion Channels/genetics , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Phosphorylation , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/physiology , Protein Conformation , Protein Folding , Protein Processing, Post-Translational/physiology , Protein Sorting Signals/physiology , Signal Transduction/physiology
13.
Am J Physiol Heart Circ Physiol ; 286(6): H2434-41, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14975928

ABSTRACT

Long QT syndrome (LQTS) is a cardiac repolarization disorder that can lead to arrhythmias and sudden death. Chromosome 7-linked inherited LQTS (LQT2) is caused by mutations in human ether-a-go-go-related gene (HERG; KCNH2), whereas drug-induced LQTS is caused primarily by HERG channel block. Many common polymorphisms are functionally silent and have been traditionally regarded as benign and without physiological consequence. However, the identification of common nonsynonymous single nucleotide polymorphisms (nSNPs; i.e., amino-acid coding variants) with functional phenotypes in the SCN5A Na(+) channel and MiRP1 K(+) channel beta-subunit have challenged this viewpoint. In this report, we test the hypothesis that common missense HERG polymorphisms alter channel physiology. Comprehensive mutational analysis of HERG was performed on genomic DNA derived from a population-based cohort of sudden infant death syndrome and two reference allele cohorts derived from 100 African American and 100 Caucasian individuals. Amino acid-encoding variants were considered common polymorphisms if they were present in at least two of the three study cohorts with an allelic frequency >0.5%. Four nSNPs were identified: K897T, P967L, R1047L, and Q1068R. Wild-type (WT) and polymorphic channels were heterologously expressed in human embryonic kidney cells, and biochemical and voltage-clamp techniques were used to characterize their functional properties. All channel types were processed similarly, but several electrophysiological differences were identified: 1) K897T current density was lower than the other polymorphic channels; 2) K897T channels activated at more negative potentials than WT and R1047L; 3) K897T and Q1068R channels inactivated and recovered from inactivation faster than WT, P967L, and R1047L channels; and 4) K897T channels showed subtle differences compared with WT channels when stimulated with an action potential waveform. In contrast to K897T and Q1068R channels, P967L and R1047L channels were electrophysiologically indistinguishable from WT channels. All HERG channels had similar sensitivity to block by cisapride. Therefore, some HERG polymorphic channels are electrophysiologically different from WT channels.


Subject(s)
Cation Transport Proteins/genetics , Long QT Syndrome/genetics , Polymorphism, Single Nucleotide , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Sudden Infant Death , Cation Transport Proteins/metabolism , Cells, Cultured , Ether-A-Go-Go Potassium Channels , Humans , Infant , Kidney/cytology , Membrane Potentials/physiology , Mutagenesis, Site-Directed , Myocardium/metabolism , Phenotype , Potassium Channels/metabolism
14.
J Biol Chem ; 278(37): 35749-54, 2003 Sep 12.
Article in English | MEDLINE | ID: mdl-12837749

ABSTRACT

Several mutations in the human ether-a-go-go-related K+ channel gene (HERG or KCNH2) cause long QT syndrome (LQT2) by reducing the intracellular transport (trafficking) of the channel protein to the cell surface. Drugs that bind to and block HERG channels (i.e. E4031) rescue the surface expression of some trafficking defective LQT2 mutations. Because these drugs potently block HERG current, their ability to correct congenital LQT is confounded by their risk of causing acquired LQT. We tested the hypothesis that pharmacological rescue can occur without HERG channel block. Thapsigargin (1 microM), a sarcoplasmic/endoplasmic reticulum Ca2+-ATPase inhibitor, rescued the surface expression of G601S, and it did so without blocking current. Thapsigargin-induced rescue and E4031-induced rescue caused complex glycosylation that was evident within 3 h of drug exposure. Disruption of the Golgi apparatus with brefeldin A prevented thapsigargin- and E4031-induced rescue of IG01S. Confocal imaging showed that G601S protein is predominantly "trapped" intracellularly and that both thapsigargin and E4031 promote its relocation to the surface membrane. We also studied two other trafficking defective LQT2 mutations. Thapsigargin rescued the C terminus mutation F805C but not N470D, whereas E4031 rescued N470D but not F805C. Other sarcoplasmic/endoplasmic reticulum Ca2+-ATPase inhibitors did not rescue G601S or F805C. 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).


Subject(s)
Cation Transport Proteins , DNA-Binding Proteins , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Thapsigargin/pharmacology , Trans-Activators , Amino Acid Substitution , Asparagine , Aspartic Acid , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Cell Nucleus/drug effects , Cell Nucleus/physiology , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Glycine , Humans , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels/metabolism , Protein Transport/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Serine , Transcriptional Regulator ERG
15.
J Neurobiol ; 55(1): 73-85, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12605460

ABSTRACT

A very large number of evolutionarily conserved potassium channels have been identified but very little is known about their function or modulation in vivo. Metamorphosis of the tobacco hornworm, Manduca sexta, is a compelling model system for such studies because it permits analysis to be conducted at the level of identified neurons whose roles in simple behaviors and endocrine regulation are known. We present here the characterization of the first ion channel to be cloned from this animal. Partial genomic sequence for Manduca sexta ether à-go-go (Mseag) and a cDNA clone encoding the Mseag open reading frame were obtained. Genomic Southern analysis indicates that Manduca contains a single member of the eag subfamily per haploid genome. When expressed in Xenopus oocytes, MsEag channels conduct a voltage-dependent, K+ selective outward current with an inactivating component that closely resembles the Drosophila eag current. Mseag transcripts were restricted to the nervous system, adult antenna, and one set of larval skeletal muscles. Steroid hormonal regulation of Mseag expression is suggested by the temporal correlation of developmental changes in transcript expression with the changing steroid titers that promote metamorphosis. These results provide the foundation for functional and modulatory studies of the Eag family of K+ channels in Manduca, which will complement the genetic analysis in Drosophila.


Subject(s)
Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium/metabolism , Amino Acid Sequence , Animals , Blotting, Southern , Drosophila , ERG1 Potassium Channel , Electric Conductivity , Electrophysiology , Ether-A-Go-Go Potassium Channels , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Gene Library , Genomic Library , Larva , Manduca , Metamorphosis, Biological , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Nervous System/growth & development , Nervous System/metabolism , Oocytes/physiology , Potassium Channels/biosynthesis , Potassium Channels/physiology , RNA, Messenger/analysis , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, Protein , Species Specificity , Steroid Hydroxylases/pharmacology , Xenopus laevis
16.
Curr Cardiol Rep ; 4(5): 411-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12169238

ABSTRACT

The recognition of the role that genetic abnormalities play in the generation of cardiac arrhythmias and sudden cardiac death has evolved enormously over the past decade. One result is new insight into underlying physiologic and pathophysiologic mechanisms. New therapies based on this evolving insight are being developed. This review summarizes recent discoveries with a focus on the genetic basis of cardiac arrhythmias and their implications for new therapies.


Subject(s)
Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/epidemiology , Arrhythmias, Cardiac/therapy , Death, Sudden, Cardiac/epidemiology , Death, Sudden, Cardiac/etiology , Genetic Therapy , Humans , Risk Factors , United States/epidemiology
17.
Circulation ; 105(24): 2830-5, 2002 Jun 18.
Article in English | MEDLINE | ID: mdl-12070109

ABSTRACT

BACKGROUND: Defective protein trafficking is a consequence of gene mutations. Human long-QT (LQT) syndrome results from mutations in several genes, including the human ether-a-go-go-related gene (HERG), which encodes a delayed rectifier K(+) current. Trafficking-defective mutant HERG protein is a mechanism for reduced delayed rectifier K(+) current in LQT2, and high-affinity HERG channel-blocking drugs can result in pharmacological rescue. Methods and Results- We postulated that drug molecules modified to remove high-affinity HERG block may still stabilize mutant proteins in a conformation required for rescue. We tested terfenadine carboxylate (fexofenadine) and terfenadine, structurally similar drugs with markedly different affinities for HERG block, for rescue of trafficking-defective LQT2 mutations. Terfenadine rescued the N470D mutation but blocked the channels. In contrast, fexofenadine rescued N470D with a half-maximal rescue concentration of 177 nmol/L, which is approximately 350-fold lower than the half-maximal channel block concentration. The G601S mutation was also rescued without channel block. CONCLUSIONS: Pharmacological rescue can occur without channel block. This could represent a new antiarrhythmic paradigm in the treatment of some trafficking-defective LQT2 mutations.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/genetics , Potassium Channels/physiology , Terfenadine/analogs & derivatives , Terfenadine/pharmacology , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Humans , Kinetics , Long QT Syndrome/genetics , Membrane Potentials , Mutation , Potassium Channel Blockers
SELECTION OF CITATIONS
SEARCH DETAIL
...