1
|
Wang X, Harty KR, Wan TC, Qu Z, Smith BC, Lough JW, Auchampach JA. Mitigation of Injury from Myocardial Infarction by Pentamidine, an Inhibitor of the Acetyltransferase Tip60. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07696-z. [PMID: 40202550 DOI: 10.1007/s10557-025-07696-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
PURPOSE There is an urgent unmet need for new pharmacologic approaches that promote re-muscularization and repair following myocardial infarction (MI). We previously reported that genetic depletion of the acetyltransferase Tip60 after MI in a mouse model activates the CM cell-cycle, reduces scarring, and restores cardiac function, and that these beneficial effects are mimicked by the Tip60-selective inhibitor TH1834. Here, we investigated whether the FDA-approved anti-microbial agent pentamidine, a Tip60 inhibitor from which TH1834 is derived, also protects from the damaging effects of MI. METHODS Adult (10-14 weeks old) C57Bl/6 mice were subjected to permanent left coronary artery ligation to induce MI. Subsequently, echocardiography, electrocardiography, cardiac staining, and molecular analyses were performed to monitor the effects of treatment with pentamidine on cardiac injury and function. RESULTS We report that transient systemic administration of pentamidine on days 3-16 post-MI at a daily dose of 3 mg/kg efficiently improved cardiac function for up to ten months. This was accompanied by improved survival, diminished scarring, and increased activation of cell-cycle markers in CMs located in the infarct border zone in the absence of hypertrophy. Histological assessments suggested that post-MI treatment with pentamidine reduced site-specific acetylation of the minor histone variant H2A.Z at lysines K4 and K7 in CMs, indicative of the dedifferentiation process which must occur prior to CM proliferation. Treating mice with pentamidine post-MI produced no prominent electrophysiological changes. CONCLUSIONS These findings support the translational potential of pentamidine for treatment of MI, and provide evidence that functional improvement is mediated, in part, by CM renewal due to inhibition of the acetyltransferase activity of Tip60.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Katherine R Harty
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Tina C Wan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Zhuocheng Qu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - John W Lough
- Department of Cell Biology Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - John A Auchampach
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
2
|
Bloothooft M, Verbruggen B, Seibertz F, van der Heyden MAG, Voigt N, de Boer TP. Recording ten-fold larger I Kr conductances with automated patch clamping using equimolar Cs + solutions. Front Physiol 2024; 15:1298340. [PMID: 38328302 PMCID: PMC10847579 DOI: 10.3389/fphys.2024.1298340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/09/2024] [Indexed: 02/09/2024] Open
Abstract
Background: The rapid delayed rectifier potassium current (IKr) is important for cardiac repolarization and is most often involved in drug-induced arrhythmias. However, accurately measuring this current can be challenging in human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes because of its small current density. Interestingly, the ion channel conducting IKr, hERG channel, is not only permeable to K+ ions but also to Cs+ ions when present in equimolar concentrations inside and outside of the cell. Methods: In this study, IhERG was measured from Chinese hamster ovary (CHO)-hERG cells and hiPSC-CM using either Cs+ or K+ as the charge carrier. Equimolar Cs+ has been used in the literature in manual patch-clamp experiments, and here, we apply this approach using automated patch-clamp systems. Four different (pre)clinical drugs were tested to compare their effects on Cs+- and K+-based currents. Results: Using equimolar Cs+ solutions gave rise to approximately ten-fold larger hERG conductances. Comparison of Cs+- and K+-mediated currents upon application of dofetilide, desipramine, moxifloxacin, or LUF7244 revealed many similarities in inhibition or activation properties of the drugs studied. Using equimolar Cs+ solutions gave rise to approximately ten-fold larger hERG conductances. In hiPSC-CM, the Cs+-based conductance is larger compared to the known K+-based conductance, and the Cs+ hERG conductance can be inhibited similarly to the K+-based conductance. Conclusion: Using equimolar Cs+ instead of K+ for IhERG measurements in an automated patch-clamp system gives rise to a new method by which, for example, quick scans can be performed on effects of drugs on hERG currents. This application is specifically relevant when such experiments are performed using cells which express small IKr current densities in combination with small membrane capacitances.
Collapse
Affiliation(s)
- Meye Bloothooft
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bente Verbruggen
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Nanion Technologies GmbH, Munich, Germany
| | - Marcel A. G. van der Heyden
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Teun P. de Boer
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
3
|
Meier S, Grundland A, Dobrev D, Volders PG, Heijman J. In silico analysis of the dynamic regulation of cardiac electrophysiology by K v 11.1 ion-channel trafficking. J Physiol 2023; 601:2711-2731. [PMID: 36752166 PMCID: PMC10313819 DOI: 10.1113/jp283976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Cardiac electrophysiology is regulated by continuous trafficking and internalization of ion channels occurring over minutes to hours. Kv 11.1 (also known as hERG) underlies the rapidly activating delayed-rectifier K+ current (IKr ), which plays a major role in cardiac ventricular repolarization. Experimental characterization of the distinct temporal effects of genetic and acquired modulators on channel trafficking and gating is challenging. Computer models are instrumental in elucidating these effects, but no currently available model incorporates ion-channel trafficking. Here, we present a novel computational model that reproduces the experimentally observed production, forward trafficking, internalization, recycling and degradation of Kv 11.1 channels, as well as their modulation by temperature, pentamidine, dofetilide and extracellular K+ . The acute effects of these modulators on channel gating were also incorporated and integrated with the trafficking model in the O'Hara-Rudy human ventricular cardiomyocyte model. Supraphysiological dofetilide concentrations substantially increased Kv 11.1 membrane levels while also producing a significant channel block. However, clinically relevant concentrations did not affect trafficking. Similarly, severe hypokalaemia reduced Kv 11.1 membrane levels based on long-term culture data, but had limited effect based on short-term data. By contrast, clinically relevant elevations in temperature acutely increased IKr due to faster kinetics, while after 24 h, IKr was decreased due to reduced Kv 11.1 membrane levels. The opposite was true for lower temperatures. Taken together, our model reveals a complex temporal regulation of cardiac electrophysiology by temperature, hypokalaemia, and dofetilide through competing effects on channel gating and trafficking, and provides a framework for future studies assessing the role of impaired trafficking in cardiac arrhythmias. KEY POINTS: Kv 11.1 channels underlying the rapidly activating delayed-rectifier K+ current are important for ventricular repolarization and are continuously shuttled from the cytoplasm to the plasma membrane and back over minutes to hours. Kv 11.1 gating and trafficking are modulated by temperature, drugs and extracellular K+ concentration but experimental characterization of their combined effects is challenging. Computer models may facilitate these analyses, but no currently available model incorporates ion-channel trafficking. We introduce a new two-state ion-channel trafficking model able to reproduce a wide range of experimental data, along with the effects of modulators of Kv 11.1 channel functioning and trafficking. The model reveals complex dynamic regulation of ventricular repolarization by temperature, extracellular K+ concentration and dofetilide through opposing acute (millisecond) effects on Kv 11.1 gating and long-term (hours) modulation of Kv 11.1 trafficking. This in silico trafficking framework provides a tool to investigate the roles of acute and long-term processes on arrhythmia promotion and maintenance.
Collapse
Affiliation(s)
- Stefan Meier
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Adaïa Grundland
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Data Science and Knowledge Engineering, Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Quebec, Canada
| | - Paul G.A. Volders
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine, and Life Sciences, Maastricht University and Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
4
|
Vedder VL, Reinberger T, Haider SMI, Eichelmann L, Odenthal N, Abdelilah-Seyfried S, Aherrahrou Z, Breuer M, Erdmann J. pyHeart4Fish: Chamber-specific heart phenotype quantification of zebrafish in high-content screens. Front Cell Dev Biol 2023; 11:1143852. [PMID: 37113769 PMCID: PMC10126419 DOI: 10.3389/fcell.2023.1143852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death. Of CVDs, congenital heart diseases are the most common congenital defects, with a prevalence of 1 in 100 live births. Despite the widespread knowledge that prenatal and postnatal drug exposure can lead to congenital abnormalities, the developmental toxicity of many FDA-approved drugs is rarely investigated. Therefore, to improve our understanding of drug side effects, we performed a high-content drug screen of 1,280 compounds using zebrafish as a model for cardiovascular analyses. Zebrafish are a well-established model for CVDs and developmental toxicity. However, flexible open-access tools to quantify cardiac phenotypes are lacking. Here, we provide pyHeart4Fish, a novel Python-based, platform-independent tool with a graphical user interface for automated quantification of cardiac chamber-specific parameters, such as heart rate (HR), contractility, arrhythmia score, and conduction score. In our study, about 10.5% of the tested drugs significantly affected HR at a concentration of 20 µM in zebrafish embryos at 2 days post-fertilization. Further, we provide insights into the effects of 13 compounds on the developing embryo, including the teratogenic effects of the steroid pregnenolone. In addition, analysis with pyHeart4Fish revealed multiple contractility defects induced by seven compounds. We also found implications for arrhythmias, such as atrioventricular block caused by chloropyramine HCl, as well as (R)-duloxetine HCl-induced atrial flutter. Taken together, our study presents a novel open-access tool for heart analysis and new data on potentially cardiotoxic compounds.
Collapse
Affiliation(s)
- Viviana L. Vedder
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Tobias Reinberger
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Syed M. I. Haider
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Luis Eichelmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Nadine Odenthal
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Salim Abdelilah-Seyfried
- Faculty of Mathematics and Natural Sciences, Institute for Biochemistry and Biology, University Potsdam, Potsdam, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| | - Maximilian Breuer
- Faculty of Mathematics and Natural Sciences, Institute for Biochemistry and Biology, University Potsdam, Potsdam, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
- University Heart Centre Lübeck, Lübeck, Germany
| |
Collapse
|
5
|
Xu X, Yin Y, Li D, Yao B, Zhao L, Wang H, Wang H, Dong J, Zhang J, Peng R. Vicious LQT induced by a combination of factors different from hERG inhibition. Front Pharmacol 2022; 13:930831. [PMID: 35935820 PMCID: PMC9354841 DOI: 10.3389/fphar.2022.930831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Clinically, drug-induced torsades de pointes (TdP) are rare events, whereas the reduction of the human ether-à-go-go-related gene (hERG) current is common. In this study, we aimed to explore the specific factors that contribute to the deterioration of hERG inhibition into malignant ventricular arrhythmias. Cisapride, a drug removed from the market because it caused long QT (LQT) syndrome and torsade de pointes (TdP), was used to induce hERG inhibition. The effects of cisapride on the hERG current were evaluated using a whole-cell patch clamp. Based on the dose-response curve of cisapride, models of its effects at different doses (10, 100, and 1,000 nM) on guinea pig heart in vitro were established. The effects of cisapride on electrocardiogram (ECG) signals and QT interval changes in the guinea pigs were then comprehensively evaluated by multi-channel electrical mapping and high-resolution fluorescence mapping, and changes in the action potential were simultaneously detected. Cisapride dose-dependently inhibited the hERG current with a half inhibitory concentration (IC50) of 32.63 ± 3.71 nM. The complete hERG suppression by a high dose of cisapride (1,000 nM) prolonged the action potential duration (APD), but not early after depolarizations (EADs) and TdP occurred. With 1 μM cisapride and lower Mg2+/K+, the APD exhibited triangulation, dispersion, and instability. VT was induced in two of 12 guinea pig hearts. Furthermore, the combined administration of isoproterenol was not therapeutic and increased susceptibility to ventricular fibrillation (VF) development. hERG inhibition alone led to QT and ERP prolongation and exerted an anti-arrhythmic effect. However, after the combination with low concentrations of magnesium and potassium, the prolonged action potential became unstable, triangular, and dispersed, and VT was easy to induce. The combination of catecholamines shortened the APD, but triangulation and dispersion still existed. At this time, VF was easily induced and sustained.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jing Zhang
- *Correspondence: Jing Zhang, ; Ruiyun Peng,
| | | |
Collapse
|
6
|
The Therapeutic Potential of Carnosine as an Antidote against Drug-Induced Cardiotoxicity and Neurotoxicity: Focus on Nrf2 Pathway. Molecules 2022; 27:molecules27144452. [PMID: 35889325 PMCID: PMC9324774 DOI: 10.3390/molecules27144452] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Different drug classes such as antineoplastic drugs (anthracyclines, cyclophosphamide, 5-fluorouracil, taxanes, tyrosine kinase inhibitors), antiretroviral drugs, antipsychotic, and immunosuppressant drugs are known to induce cardiotoxic and neurotoxic effects. Recent studies have demonstrated that the impairment of the nuclear factor erythroid 2–related factor 2 (Nrf2) pathway is a primary event in the pathophysiology of drug-induced cardiotoxicity and neurotoxicity. The Nrf2 pathway regulates the expression of different genes whose products are involved in antioxidant and inflammatory responses and the detoxification of toxic species. Cardiotoxic drugs, such as the anthracycline doxorubicin, or neurotoxic drugs, such as paclitaxel, suppress or impair the Nrf2 pathway, whereas the rescue of this pathway counteracts both the oxidative stress and inflammation that are related to drug-induced cardiotoxicity and neurotoxicity. Therefore Nrf2 represents a novel pharmacological target to develop new antidotes in the field of clinical toxicology. Interestingly, carnosine (β-alanyl-l-histidine), an endogenous dipeptide that is characterized by strong antioxidant, anti-inflammatory, and neuroprotective properties is able to rescue/activate the Nrf2 pathway, as demonstrated by different preclinical studies and preliminary clinical evidence. Starting from these new data, in the present review, we examined the evidence on the therapeutic potential of carnosine as an endogenous antidote that is able to rescue the Nrf2 pathway and then counteract drug-induced cardiotoxicity and neurotoxicity.
Collapse
|
7
|
MacNair CR, Farha MA, Serrano-Wu MH, Lee KK, Hubbard B, Côté JP, Carfrae LA, Tu MM, Gaulin JL, Hunt DK, Hung DT, Brown ED. Preclinical Development of Pentamidine Analogs Identifies a Potent and Nontoxic Antibiotic Adjuvant. ACS Infect Dis 2022; 8:768-777. [PMID: 35319198 DOI: 10.1021/acsinfecdis.1c00482] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The difficulty in treating Gram-negative bacteria can largely be attributed to their highly impermeable outer membrane (OM), which serves as a barrier to many otherwise active antibiotics. This can be overcome with the use of perturbant molecules, which disrupt OM integrity and sensitize Gram-negative bacteria to many clinically available Gram-positive-active antibiotics. Although many new perturbants have been identified in recent years, most of these molecules are impeded by toxicity due to the similarities between pathogen and host cell membranes. For example, our group recently reported the cryptic OM-perturbing activity of the antiprotozoal drug pentamidine. Its development as an antibiotic adjuvant is limited, however, by toxicity concerns. Herein, we took a medicinal chemistry approach to develop novel analogs of pentamidine, aiming to improve its OM activity while reducing its off-target toxicity. We identified the compound P35, which induces OM disruption and potentiates Gram-positive-active antibiotics in Acinetobacter baumannii and Klebsiella pneumoniae. Relative to pentamidine, P35 has reduced mammalian cell cytotoxicity and hERG trafficking inhibition. Additionally, P35 outperforms pentamidine in a murine model of A. baumannii bacteremia. Together, this preclinical analysis supports P35 as a promising lead for further development as an OM perturbant.
Collapse
Affiliation(s)
- Craig R. MacNair
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Maya A. Farha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Michael H. Serrano-Wu
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Katie K. Lee
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Brian Hubbard
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jean-Philippe Côté
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Lindsey A. Carfrae
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Megan M. Tu
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Jeffrey L. Gaulin
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Diana K. Hunt
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Deborah T. Hung
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| |
Collapse
|
8
|
Mamoshina P, Rodriguez B, Bueno-Orovio A. Toward a broader view of mechanisms of drug cardiotoxicity. CELL REPORTS MEDICINE 2021; 2:100216. [PMID: 33763655 PMCID: PMC7974548 DOI: 10.1016/j.xcrm.2021.100216] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiotoxicity, defined as toxicity that affects the heart, is one of the most common adverse drug effects. Numerous drugs have been shown to have the potential to induce lethal arrhythmias by affecting cardiac electrophysiology, which is the focus of current preclinical testing. However, a substantial number of drugs can also affect cardiac function beyond electrophysiology. Within this broader sense of cardiotoxicity, this review discusses the key drug-protein interactions known to be involved in cardiotoxic drug response. We cover adverse effects of anticancer, central nervous system, genitourinary system, gastrointestinal, antihistaminic, anti-inflammatory, and anti-infective agents, illustrating that many share mechanisms of cardiotoxicity, including contractility, mitochondrial function, and cellular signaling.
Collapse
Affiliation(s)
| | - Blanca Rodriguez
- Department of Computer Science, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Alfonso Bueno-Orovio
- Department of Computer Science, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Detection and impact of hysteresis when evaluating a drug's QTc effect using concentration-QTc analysis. J Pharmacokinet Pharmacodyn 2020; 48:187-202. [PMID: 33118135 DOI: 10.1007/s10928-020-09725-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/14/2020] [Indexed: 10/23/2022]
Abstract
Early-phase studies quantifying the QTc prolongation potential for a new drug often use linear concentration-QTc (C-QTc) models, assuming no delay between plasma concentrations and QTc changes. However, that assumption is not always correct. The term "hysteresis" has been utilized to describe a time lag present between a measurable concentration and a measurable effect. To detect and quantify hysteresis and its impact on study interpretation, studies with hysteresis of 0.25-4 h were simulated with different doses, half-lives, and sampling schedules in a crossover design. Hysteresis was quantified using a novel method termed exposure-normalized GRI (enGRI), a proposed modification of the Glomb-Ring Index (GRI), to account for delay and magnitude of QTc effects. With realistic sampling, the rate of false negative studies (FN) increased proportionally to the delay, even for delays shorter than 1 h. Using an enGRI threshold (γ) of 2 ms resulted in FN with undetected delay and FN without hysteresis at approximately the same rate. For γ = 2 ms, the specificity of enGRI was > 90% throughout the investigated scenarios. We therefore propose the incorporation of enGRI when interpreting results from C-QTc analysis with the intent of characterizing QTc effects.
Collapse
|
10
|
Shi YQ, Fan P, Zhang GC, Zhang YH, Li MZ, Wang F, Li BX. Probucol-induced hERG Channel Reduction can be Rescued by Matrine and Oxymatrine in vitro. Curr Pharm Des 2020; 25:4606-4612. [PMID: 31657676 PMCID: PMC7327797 DOI: 10.2174/1381612825666191026170033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/19/2019] [Indexed: 01/24/2023]
Abstract
Background The human ether-a-go-go-related gene (hERG) potassium channel is the rapidly activating component of cardiac delayed rectifier potassium current (IKr), which is a crucial determinant of cardiac repolarization. The reduction of hERG current is commonly believed to cause Long QT Syndrome (LQTs). Probucol, a cholesterol-lowering drug, induces LQTs by inhibiting the expression of the hERG channel. Unfortunately, there is currently no effective therapeutic method to rescue probucol-induced LQTs. Methods Patch-clamp recording techniques were used to detect the action potential duration (APD) and current of hERG. Western blot was performed to measure the expression levels of proteins. Results In this study, we demonstrated that 1 μM matrine and oxymatrine could rescue the hERG current and hERG surface expression inhibited by probucol. In addition, matrine and oxymatrine significantly shortened the prolonged action potential duration induced by probucol in neonatal cardiac myocytes. We proposed a novel mechanism underlying the probucol induced decrease in the expression of transcription factor Specificity protein 1 (Sp1), which is an established transactivator of the hERG gene. We also demonstrated that matrine and oxymatrine were able to upregulate Sp1 expression which may be one of the possible mechanisms by which matrine and oxymatrine rescued probucol-induced hERG channel deficiency. Conclusion Our current results demonstrate that matrine and oxymatrine could rescue probucol-induced hERG deficiency in vitro, which may lead to potentially effective therapeutic drugs for treating acquired LQT2 by probucol in the future.
Collapse
Affiliation(s)
- Yuan-Qi Shi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, China
| | - Pan Fan
- Department of Ophthalmology, the Second Affiliated Hospital of Harbin Medical University, No. 148 Baojian Road, Nangang District, Harbin 150081, China
| | - Guo-Cui Zhang
- Department of Pharmacology, Harbin Medical University, No. 157 Baojian Road, Harbin, 150086, China
| | - Yu-Hao Zhang
- Department of Pharmacology, Harbin Medical University, No. 157 Baojian Road, Harbin, 150086, China
| | - Ming-Zhu Li
- Department of Pharmacology, Harbin Medical University, No. 157 Baojian Road, Harbin, 150086, China
| | - Fang Wang
- Department of Pharmacology, Harbin Medical University, No. 157 Baojian Road, Harbin, 150086, China
| | - Bao-Xin Li
- Department of Pharmacology, Harbin Medical University, No. 157 Baojian Road, Harbin, 150086, China.,State-Province Key Laboratory of Biopharmaceutical Engineering, No. 157 Baojian Road, Harbin, 150086, China
| |
Collapse
|
11
|
Qile M, Ji Y, Golden TD, Houtman MJC, Romunde F, Fransen D, van Ham WB, IJzerman AP, January CT, Heitman LH, Stary-Weinzinger A, Delisle BP, van der Heyden MAG. LUF7244 plus Dofetilide Rescues Aberrant K v11.1 Trafficking and Produces Functional I Kv11.1. Mol Pharmacol 2020; 97:355-364. [PMID: 32241959 DOI: 10.1124/mol.119.118190] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/24/2020] [Indexed: 02/14/2025] Open
Abstract
Voltage-gated potassium 11.1 (Kv11.1) channels play a critical role in repolarization of cardiomyocytes during the cardiac action potential (AP). Drug-mediated Kv11.1 blockade results in AP prolongation, which poses an increased risk of sudden cardiac death. Many drugs, like pentamidine, interfere with normal Kv11.1 forward trafficking and thus reduce functional Kv11.1 channel densities. Although class III antiarrhythmics, e.g., dofetilide, rescue congenital and acquired forward trafficking defects, this is of little use because of their simultaneous acute channel blocking effect. We aimed to test the ability of a combination of dofetilide plus LUF7244, a Kv11.1 allosteric modulator/activator, to rescue Kv11.1 trafficking and produce functional Kv11.1 current. LUF7244 treatment by itself did not disturb or rescue wild type (WT) or G601S-Kv11.1 trafficking, as shown by Western blot and immunofluorescence microcopy analysis. Pentamidine-decreased maturation of WT Kv11.1 levels was rescued by 10 μM dofetilide or 10 μM dofetilide + 5 μM LUF7244. In trafficking defective G601S-Kv11.1 cells, dofetilide (10 μM) or dofetilide + LUF7244 (10 + 5 μM) also restored Kv11.1 trafficking, as demonstrated by Western blot and immunofluorescence microscopy. LUF7244 (10 μM) increased IKv 11.1 despite the presence of dofetilide (1 μM) in WT Kv11.1 cells. In G601S-expressing cells, long-term treatment (24-48 hour) with LUF7244 (10 μM) and dofetilide (1 μM) increased IKv11.1 compared with nontreated or acutely treated cells. We conclude that dofetilide plus LUF7244 rescues Kv11.1 trafficking and produces functional IKv11.1 Thus, combined administration of LUF7244 and an IKv11.1 trafficking corrector could serve as a new pharmacological therapy of both congenital and drug-induced Kv11.1 trafficking defects. SIGNIFICANCE STATEMENT: Decreased levels of functional Kv11.1 potassium channel at the plasma membrane of cardiomyocytes prolongs action potential repolarization, which associates with cardiac arrhythmia. Defective forward trafficking of Kv11.1 channel protein is an important factor in acquired and congenital long QT syndrome. LUF7244 as a negative allosteric modulator/activator in combination with dofetilide corrected both congenital and acquired Kv11.1 trafficking defects, resulting in functional Kv11.1 current.
Collapse
Affiliation(s)
- Muge Qile
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Yuan Ji
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Tyona D Golden
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Marien J C Houtman
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Fee Romunde
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Doreth Fransen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Willem B van Ham
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Ad P IJzerman
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Craig T January
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Laura H Heitman
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Anna Stary-Weinzinger
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Brian P Delisle
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| | - Marcel A G van der Heyden
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands (M.Q., Y.J., M.J.C.H., F.R., D.F., W.B.H., M.A.G.H.); Department of Physiology, University of Kentucky, Lexington, Kentucky (T.D.G., B.P.D.); Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (W.B.H., A.S.-W.); Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, The Netherlands (A.P.I., L.H.H.); and Department of Medicine, University of Wisconsin, Madison, Wisconsin (C.T.J.)
| |
Collapse
|
12
|
Tay YL, Amanah A, Adenan MI, Wahab HA, Tan ML. Mitragynine, an euphoric compound inhibits hERG1a/1b channel current and upregulates the complexation of hERG1a-Hsp90 in HEK293-hERG1a/1b cells. Sci Rep 2019; 9:19757. [PMID: 31874991 PMCID: PMC6930223 DOI: 10.1038/s41598-019-56106-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 11/06/2019] [Indexed: 11/24/2022] Open
Abstract
Mitragyna speciosa Korth (M. speciosa) has been widely used as a recreational product, however, there are growing concerns on the abuse potentials and toxicity of the plant. Several poisoning and fatal cases involving kratom and mitragynine have been reported but the underlying causes remain unclear. The human ether-a-go-go-related gene 1 (hERG1) encodes the pore-forming subunit underlying cardiac rapidly delayed rectifier potassium current (IKr). Pharmacological blockade of the IKr can cause acquired long QT syndrome, leading to lethal cardiac arrhythmias. This study aims to elucidate the mechanisms of mitragynine-induced inhibition on hERG1a/1b current. Electrophysiology experiments were carried out using Port-a-Patch system. 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 was found to inhibit the IKr current with an IC50 value of 332.70 nM. It causes a significant reduction of the fully-glycosylated (fg) hERG1a protein expression but upregulates both core-glycosylated (cg) expression and hERG1a-Hsp90 complexes, suggesting possible impaired hERG1a trafficking. In conclusion, mitragynine inhibits hERG1a/1b current through direct channel blockade at lower concentration, but at higher concentration, it upregulates the complexation of hERG1a-Hsp90 which may be inhibitory towards channel trafficking.
Collapse
Affiliation(s)
- Yea Lu Tay
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, NIBM, Ministry of Energy, Science, Technology, Environment and Climate Change (MESTECC), Pulau Pinang, 11700, Malaysia
| | - Azimah Amanah
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, NIBM, Ministry of Energy, Science, Technology, Environment and Climate Change (MESTECC), Pulau Pinang, 11700, Malaysia
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Selangor Darul Ehsan, 42300, Malaysia
| | - Habibah Abdul Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, 11700, Malaysia
| | - Mei Lan Tan
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, NIBM, Ministry of Energy, Science, Technology, Environment and Climate Change (MESTECC), Pulau Pinang, 11700, Malaysia. .,School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, 11700, Malaysia. .,Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, Kepala Batas, Pulau Pinang, 13200, Malaysia.
| |
Collapse
|
13
|
Lamore SD, Kohnken RA, Peters MF, Kolaja KL. Cardiovascular Toxicity Induced by Kinase Inhibitors: Mechanisms and Preclinical Approaches. Chem Res Toxicol 2019; 33:125-136. [DOI: 10.1021/acs.chemrestox.9b00387] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sarah D. Lamore
- Preclinical Development, Wave Life Sciences, Lexington, Massachusetts 02421, United States
| | - Rebecca A. Kohnken
- Preclinical Safety, Abbvie, North Chicago, Illinois 60064, United States
| | - Matthew F. Peters
- Oncology Safety, Clinical Pharmacology and Safety Sciences, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, United States
| | - Kyle L. Kolaja
- Investigative Toxicology and Cell Therapy Safety, Nonclinical Development, Celgene Corporation, Summit, New Jersey 07901, United States
| |
Collapse
|
14
|
Dong X, Liu Y, Niu H, Wang G, Dong L, Zou A, Wang K. Electrophysiological characterization of a small molecule activator on human ether-a-go-go-related gene (hERG) potassium channel. J Pharmacol Sci 2019; 140:284-290. [DOI: 10.1016/j.jphs.2019.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/02/2023] Open
|
15
|
Goversen B, Jonsson MK, van den Heuvel NH, Rijken R, Vos MA, van Veen TA, de Boer TP. The influence of hERG1a and hERG1b isoforms on drug safety screening in iPSC-CMs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 149:86-98. [PMID: 30826123 DOI: 10.1016/j.pbiomolbio.2019.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/14/2019] [Accepted: 02/08/2019] [Indexed: 01/03/2023]
Abstract
The human Ether-à-go-go Related Gene (hERG) encodes the pore forming subunit of the channel that conducts the rapid delayed rectifier potassium current IKr. IKr drives repolarization in the heart and when IKr is dysfunctional, cardiac repolarization delays, the QT interval on the electrocardiogram (ECG) prolongs and the risk of developing lethal arrhythmias such as Torsade de Pointes (TdP) increases. TdP risk is incorporated in drug safety screening for cardiotoxicity where hERG is the main target since the IKr channels appear highly sensitive to blockage. hERG block is also included as an important read-out in the Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative which aims to combine in vitro and in silico experiments on induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) to screen for cardiotoxicity. However, the hERG channel has some unique features to consider for drug safety screening, which we will discuss in this study. The hERG channel consists of different isoforms, hERG1a and hERG1b, which individually influence the kinetics of the channel and the drug response in the human heart and in iPSC-CMs. hERG1b is often underappreciated in iPSC-CM studies, drug screening assays and in silico models, and the fact that its contribution might substantially differ between iPSC-CM and healthy but also diseased human heart, adds to this problem. In this study we show that the activation kinetics in iPSC-CMs resemble hERG1b kinetics using Cs+ as a charge carrier. Not including hERG1b in drug safety testing might underestimate the actual role of hERG1b in repolarization and drug response, and might lead to inappropriate conclusions. We stress to focus more on including hERG1b in drug safety testing concerning IKr.
Collapse
Affiliation(s)
- Birgit Goversen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands
| | - Malin Kb Jonsson
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands; Bioscience Heart Failure, Cardiovascular, Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Nikki Hl van den Heuvel
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands
| | - Rianne Rijken
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands
| | - Marc A Vos
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands
| | - Toon Ab van Veen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, the Netherlands.
| |
Collapse
|
16
|
Żołek T, Qile M, Kaźmierczak P, Bloothooft M, van der Heyden MAG, Maciejewska D. Drug-likeness of linear pentamidine analogues and their impact on the hERG K+channel – correlation with structural features. RSC Adv 2019; 9:38355-38371. [PMID: 35540224 PMCID: PMC9082326 DOI: 10.1039/c9ra08404e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/15/2019] [Indexed: 01/08/2023] Open
Abstract
The pentamidines with S atoms or sulfanilide groups in the linker have favorable drug-likeness parameters and low toxicity.
Collapse
Affiliation(s)
- Teresa Żołek
- Department of Organic Chemistry
- Faculty of Pharmacy
- Medical University of Warsaw
- 02-097 Warsaw
- Poland
| | - Muge Qile
- Department of Medical Physiology
- Division Heart & Lungs
- University Medical Center Utrecht
- Utrecht
- The Netherlands
| | - Paweł Kaźmierczak
- Department of Organic Chemistry
- Faculty of Pharmacy
- Medical University of Warsaw
- 02-097 Warsaw
- Poland
| | - Meye Bloothooft
- Department of Medical Physiology
- Division Heart & Lungs
- University Medical Center Utrecht
- Utrecht
- The Netherlands
| | - Marcel A. G. van der Heyden
- Department of Medical Physiology
- Division Heart & Lungs
- University Medical Center Utrecht
- Utrecht
- The Netherlands
| | - Dorota Maciejewska
- Department of Organic Chemistry
- Faculty of Pharmacy
- Medical University of Warsaw
- 02-097 Warsaw
- Poland
| |
Collapse
|
17
|
Asahi Y, Nomura F, Abe Y, Doi M, Sakakura T, Takasuna K, Yasuda K. Electrophysiological evaluation of pentamidine and 17-AAG in human stem cell-derived cardiomyocytes for safety assessment. Eur J Pharmacol 2019; 842:221-230. [DOI: 10.1016/j.ejphar.2018.10.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 10/28/2022]
|
18
|
Cheung SYA, Parkinson J, Wählby-Hamrén U, Dota CD, Kragh ÅM, Bergenholm L, Vik T, Collins T, Arfvidsson C, Pollard CE, Tomkinson HK, Hamrén B. A tutorial on model informed approaches to cardiovascular safety with focus on cardiac repolarisation. J Pharmacokinet Pharmacodyn 2018; 45:365-381. [PMID: 29736890 DOI: 10.1007/s10928-018-9589-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Drugs can affect the cardiovascular (CV) system either as an intended treatment or as an unwanted side effect. In both cases, drug-induced cardiotoxicities such as arrhythmia and unfavourable hemodynamic effects can occur, and be described using mathematical models; such a model informed approach can provide valuable information during drug development and can aid decision-making. However, in order to develop informative models, it is vital to understand CV physiology. The aims of this tutorial are to present (1) key background biological and medical aspects of the CV system, (2) CV electrophysiology, (3) CV safety concepts, (4) practical aspects of development of CV models and (5) regulatory expectations with a focus on using model informed and quantitative approaches to support nonclinical and clinical drug development. In addition, we share several case studies to provide practical information on project strategy (planning, key questions, assumptions setting, and experimental design) and mathematical models development that support decision-making during drug discovery and development.
Collapse
Affiliation(s)
- S Y A Cheung
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| | - J Parkinson
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - U Wählby-Hamrén
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - C D Dota
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Å M Kragh
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - L Bergenholm
- DMPK CVRM Modelling and Simulation, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - T Vik
- Cardiovascular Safety Center of Excellence, Global Medicine Development, Gothenburg, Sweden
| | - T Collins
- Safety and ADME Translational Sciences Department, Drug Safety and Metabolism, IMED Biotech Unit, Cambridge, UK
| | - C Arfvidsson
- Clinical Operation, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - C E Pollard
- Safety and ADME Translational Sciences Department, Drug Safety and Metabolism, IMED Biotech Unit, Cambridge, UK
| | - H K Tomkinson
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - B Hamrén
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
19
|
Abstract
Pharmacological chaperones (PCs) are small molecules that bind to nascent protein targets to facilitate their biogenesis. The ability of PCs to assist in the folding and subsequent forward trafficking of disease-causative protein misfolding mutants has opened new avenues for the treatment of conformational diseases such as cystic fibrosis and lysosomal storage disorders. In this chapter, an overview of the use of PCs for the treatment of conformational disorders is provided. Beyond the therapeutic application of PCs for the treatment of these disorders, pharmacological chaperoning of wild-type integral membrane proteins is discussed. Central to this discussion is the notion that the endoplasmic reticulum is a reservoir of viable but inefficiently processed wild-type protein folding intermediates whose biogenesis can be facilitated by PCs to increase functional pools. To date, the potential therapeutic use of PCs to enhance the biogenesis of wild-type proteins has received little attention. Here the rationale for the development of PCs that target WT proteins is discussed. Also considered is the likelihood that some commonly used therapeutic agents may exert unrecognized pharmacological chaperoning activity on wild-type targets in patient populations.
Collapse
Affiliation(s)
- Nancy J Leidenheimer
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.
| |
Collapse
|
20
|
Bridges RJ, Bradbury NA. Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator and Drugs: Insights from Cellular Trafficking. Handb Exp Pharmacol 2018; 245:385-425. [PMID: 29460152 DOI: 10.1007/164_2018_103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The eukaryotic cell is organized into membrane-delineated compartments that are characterized by specific cadres of proteins sustaining biochemically distinct cellular processes. The appropriate subcellular localization of proteins is key to proper organelle function and provides a physiological context for cellular processes. Disruption of normal trafficking pathways for proteins is seen in several genetic diseases, where a protein's absence for a specific subcellular compartment leads to organelle disruption, and in the context of an individual, a disruption of normal physiology. Importantly, several drug therapies can also alter protein trafficking, causing unwanted side effects. Thus, a deeper understanding of trafficking pathways needs to be appreciated as novel therapeutic modalities are proposed. Despite the promising efficacy of novel therapeutic agents, the intracellular bioavailability of these compounds has proved to be a potential barrier, leading to failures in treatments for various diseases and disorders. While endocytosis of drug moieties provides an efficient means of getting material into cells, the subsequent release and endosomal escape of materials into the cytosol where they need to act has been a barrier. An understanding of cellular protein/lipid trafficking pathways has opened up strategies for increasing drug bioavailability. Approaches to enhance endosomal exit have greatly increased the cytosolic bioavailability of drugs and will provide a means of investigating previous drugs that may have been shelved due to their low cytosolic concentration.
Collapse
Affiliation(s)
- Robert J Bridges
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA.
| |
Collapse
|
21
|
Yu D, Lv L, Fang L, Zhang B, Wang J, Zhan G, Zhao L, Zhao X, Li B. Inhibitory effects and mechanism of dihydroberberine on hERG channels expressed in HEK293 cells. PLoS One 2017; 12:e0181823. [PMID: 28763460 PMCID: PMC5538702 DOI: 10.1371/journal.pone.0181823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/09/2017] [Indexed: 12/20/2022] Open
Abstract
The human ether-a-go-go-related gene (hERG) potassium channel conducts rapid delayed rectifier potassium currents (IKr) and contributes to phase III cardiac action potential repolarization. Drugs inhibit hERG channels by binding to aromatic residues in hERG helixes. Berberine (BBR) has multiple actions, and its hydrogenated derivative dihydroberberine (DHB) is a potential candidate for developing new drugs. Previous studies have demonstrated that BBR blocks hERG channels and prolongs action potential duration (APD). Our present study aimed to investigate the effects and mechanism of DHB on hERG channels. Protein expression and the hERG current were analyzed using western blotting and patch-clamp, respectively. DHB inhibited the hERG current concentration-dependently after instantaneous perfusion, accelerated channel inactivation by directly binding tyrosine (Tyr652) and phenylalanine (Phe656), and decreased mature (155-kDa) and simultaneously increased immature (135-kDa) hERG expression, respectively. This suggests disruption of forward trafficking of hERG channels. Besides, DHB remarkably reduced heat shock protein 90 (Hsp90) expression and its interaction with hERG, indicating that DHB disrupted hERG trafficking by impairing channel folding. Meanwhie, DHB enhanced the expression of cleaved activating transcription factor-6 (ATF-6), a biomarker of unfolded protein response (UPR). Expression of calnexin and calreticulin, chaperones activated by ATF-6 to facilitate channel folding, were also increased, which indicating UPR activation. Additionally, the degradation rate of mature 155-kDa hERG increased following DHB exposure. In conclusion, we demonstrated that DHB acutely blocked hERG channels by binding the aromatic Tyr652 and Phe656. DHB may decrease hERG plasma membrane expression through two pathways involving disruption of forward trafficking of immature hERG channels and enhanced degradation of mature hERG channels. Furthermore, forward trafficking was disrupted by impaired channel folding associated with altered interactions between hERG proteins and chaperones. Finally, trafficking inhibition activated UPR, and mature hERG channel degradation was increased by DHB.
Collapse
Affiliation(s)
- Dahai Yu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Lin Lv
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Li Fang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Bo Zhang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Junnan Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Ge Zhan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Lei Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xin Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| | - Baoxin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, People's Republic of China
| |
Collapse
|
22
|
Teah YF, Abduraman MA, Amanah A, Adenan MI, Sulaiman SF, Tan ML. The effects of deoxyelephantopin on the cardiac delayed rectifier potassium channel current (I Kr) and human ether-a-go-go-related gene (hERG) expression. Food Chem Toxicol 2017; 107:293-301. [PMID: 28689918 DOI: 10.1016/j.fct.2017.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/28/2017] [Accepted: 07/04/2017] [Indexed: 12/30/2022]
Abstract
Elephantopus scaber Linn and its major bioactive component, deoxyelephantopin are known for their medicinal properties and are often reported to have various cytotoxic and antitumor activities. This plant is widely used as folk medicine for a plethora of indications although its safety profile remains unknown. Human ether-a-go-go-related gene (hERG) encodes the cardiac IKr current which is a determinant of the duration of ventricular action potentials and QT interval. The hERG potassium channel is an important antitarget in cardiotoxicity evaluation. This study investigated the effects of deoxyelephantopin on the current, mRNA and protein expression of hERG channel in hERG-transfected HEK293 cells. The hERG tail currents following depolarization pulses were insignificantly affected by deoxyelephantopin in the transfected cell line. Current reduction was less than 40% as compared with baseline at the highest concentration of 50 μM. The results were consistent with the molecular docking simulation and hERG surface protein expression. Interestingly, it does not affect the hERG expression at both transcriptional and translational level at most concentrations, although higher concentration at 10 μM caused protein accumulation. In conclusion, deoxyelephantopin is unlikely a clinically significant hERG channel and Ikr blocker.
Collapse
Affiliation(s)
- Yi Fan Teah
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, National Institutes of Biotechnology Malaysia (NIBM), Ministry of Science, Technology and Innovation Malaysia, Pulau Pinang, Malaysia
| | | | - Azimah Amanah
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, National Institutes of Biotechnology Malaysia (NIBM), Ministry of Science, Technology and Innovation Malaysia, Pulau Pinang, Malaysia
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Selangor Darul Ehsan, Malaysia
| | | | - Mei Lan Tan
- Malaysian Institute of Pharmaceuticals & Nutraceuticals, National Institutes of Biotechnology Malaysia (NIBM), Ministry of Science, Technology and Innovation Malaysia, Pulau Pinang, Malaysia; Advanced Medical & Dental Institute, Universiti Sains Malaysia, Pulau Pinang, Malaysia.
| |
Collapse
|
23
|
Stereoselective Blockage of Quinidine and Quinine in the hERG Channel and the Effect of Their Rescue Potency on Drug-Induced hERG Trafficking Defect. Int J Mol Sci 2016; 17:ijms17101648. [PMID: 27690007 PMCID: PMC5085681 DOI: 10.3390/ijms17101648] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/03/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023] Open
Abstract
Diastereoisomers of quinidine and quinine are used to treat arrhythmia and malaria, respectively. It has been reported that both drugs block the hERG (human ether-a-go-go-related gene) potassium channel which is essential for myocardium repolarization. Abnormality of repolarization increases risk of arrhythmia. The aim of our research is to study and compare the impacts of quinidine and quinine on hERG. Results show that both drugs block the hERG channel, with quinine 14-fold less potent than quinidine. In addition, they presented distinct impacts on channel dynamics. The results imply their stereospecific block effect on the hERG channel. However, F656C-hERG reversed this stereoselectivity. The mutation decreases affinity of the two drugs with hERG, and quinine was more potent than quinidine in F656C-hERG blockage. These data suggest that F656 residue contributes to the stereoselective pocket for quinidine and quinine. Further study demonstrates that both drugs do not change hERG protein levels. In rescue experiments, we found that they exert no reverse effect on pentamidine- or desipramine-induced hERG trafficking defect, although quinidine has been reported to rescue trafficking-deficient pore mutation hERG G601S based on the interaction with F656. Our research demonstrated stereoselective effects of quinidine and quinine on the hERG channel, and this is the first study to explore their reversal potency on drug-induced hERG deficiency.
Collapse
|
24
|
Mitragynine and its potential blocking effects on specific cardiac potassium channels. Toxicol Appl Pharmacol 2016; 305:22-39. [PMID: 27260674 DOI: 10.1016/j.taap.2016.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 02/07/2023]
Abstract
Mitragyna speciosa Korth is known for its euphoric properties and is frequently used for recreational purposes. Several poisoning and fatal cases involving mitragynine have been reported but the underlying causes remain unclear. Human ether-a-go-go-related gene (hERG) encodes the cardiac IKr current which is a determinant of the duration of ventricular action potentials and QT interval. On the other hand, IK1, a Kir current mediated by Kir2.1 channel and IKACh, a receptor-activated Kir current mediated by GIRK channel are also known to be important in maintaining the cardiac function. This study investigated the effects of mitragynine on the current, mRNA and protein expression of hERG channel in hERG-transfected HEK293 cells and Xenopus oocytes. The effects on Kir2.1 and GIRK channels currents were also determined in the oocytes. The hERG tail currents following depolarization pulses were inhibited by mitragynine with an IC50 value of 1.62μM and 1.15μM in the transfected cell line and Xenopus oocytes, respectively. The S6 point mutations of Y652A and F656A attenuated the inhibitor effects of mitragynine, indicating that mitragynine interacts with these high affinity drug-binding sites in the hERG channel pore cavity which was consistent with the molecular docking simulation. Interestingly, mitragynine does not affect the hERG expression at the transcriptional level but inhibits the protein expression. Mitragynine is also found to inhibit IKACh current with an IC50 value of 3.32μM but has no significant effects on IK1. Blocking of both hERG and GIRK channels may cause additive cardiotoxicity risks.
Collapse
|
25
|
Safety pharmacology studies using EFP and impedance. J Pharmacol Toxicol Methods 2016; 81:223-32. [PMID: 27084108 DOI: 10.1016/j.vascn.2016.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 12/17/2022]
Abstract
INTRODUCTION While extracellular field potential (EFP) recordings using multi-electrode arrays (MEAs) are a well-established technique for monitoring changes in cardiac and neuronal function, impedance is a relatively unexploited technology. The combination of EFP, impedance and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has important implications for safety pharmacology as functional information about contraction and field potentials can be gleaned from human cardiomyocytes in a beating monolayer. The main objectives of this study were to demonstrate, using a range of different compounds, that drug effects on contraction and electrophysiology can be detected using a beating monolayer of hiPSC-CMs on the CardioExcyte 96. METHODS hiPSC-CMs were grown as a monolayer on NSP-96 plates for the CardioExcyte 96 (Nanion Technologies) and recordings were made in combined EFP and impedance mode at physiological temperature. The effect of the hERG blockers, E4031 and dofetilide, hERG trafficking inhibitor, pentamidine, β-adrenergic receptor agonist, isoproterenol, and calcium channel blocker, nifedipine, was tested on the EFP and impedance signals. RESULTS Combined impedance and EFP measurements were made from hiPSC-CMs using the CardioExcyte 96 (Nanion Technologies). E4031 and dofetilide, known to cause arrhythmia and Torsades de Pointes (TdP) in humans, decreased beat rate in impedance and EFP modes. Early afterdepolarization (EAD)-like events, an in vitro marker of TdP, could also be detected using this system. Isoproterenol and nifedipine caused an increase in beat rate. A long-term study (over 30h) of pentamidine, a hERG trafficking inhibitor, showed a concentration and time-dependent effect of pentamidine. DISCUSSION In the light of the new Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative to improve guidelines and standardize assays and protocols, the use of EFP and impedance measurements from hiPSCs may become critical in determining the proarrhythmic risk of potential drug candidates. The combination of EFP offering information about cardiac electrophysiology, and impedance, providing information about contractility from the same area of a synchronously beating monolayer of human cardiomyocytes in a 96-well plate format has important implications for future cardiac safety testing.
Collapse
|
26
|
Smith JL, Anderson CL, Burgess DE, Elayi CS, January CT, Delisle BP. Molecular pathogenesis of long QT syndrome type 2. J Arrhythm 2016; 32:373-380. [PMID: 27761161 PMCID: PMC5063260 DOI: 10.1016/j.joa.2015.11.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/21/2015] [Accepted: 11/17/2015] [Indexed: 12/26/2022] Open
Abstract
The molecular mechanisms underlying congenital long QT syndrome (LQTS) are now beginning to be understood. New insights into the etiology and therapeutic strategies are emerging from heterologous expression studies of LQTS-linked mutant proteins, as well as inducible pluripotent stem cell derived cardiomyocytes (iPSC-CMs) from LQTS patients. This review focuses on the major molecular mechanism that underlies LQTS type 2 (LQT2). LQT2 is caused by loss of function (LOF) mutations in KCNH2 (also known as the human Ether-à-go-go-Related Gene or hERG). Most LQT2-linked mutations are missense mutations and functional studies suggest that ~90% of them disrupt the intracellular transport (trafficking) of KCNH2-encoded Kv11.1 proteins to the cell membrane. Trafficking deficient LQT2 mutations disrupt Kv11.1 protein folding and misfolded Kv11.1 proteins are retained in the endoplasmic reticulum (ER) until they are degraded in the ER associated degradation pathway (ERAD). This review focuses on the quality control mechanisms in the ER that contribute to the folding and ERAD of Kv11.1 proteins; the mechanism for ER export of Kv11.1 proteins in the secretory pathway; different subclasses of trafficking deficient LQT2 mutations; and strategies being developed to mitigate or correct trafficking deficient LQT2-related phenotypes.
Collapse
Affiliation(s)
- Jennifer L Smith
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Corey L Anderson
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Don E Burgess
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Claude S Elayi
- Department of Cardiology, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Craig T January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI, USA
| | - Brian P Delisle
- Department of Physiology, Cardiovascular Research Center, Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
27
|
Cardiotoxicity screening: a review of rapid-throughput in vitro approaches. Arch Toxicol 2015; 90:1803-16. [PMID: 26676948 DOI: 10.1007/s00204-015-1651-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/18/2015] [Indexed: 01/07/2023]
Abstract
Cardiac toxicity represents one of the leading causes of drug failure along different stages of drug development. Multiple very successful pharmaceuticals had to be pulled from the market or labeled with strict usage warnings due to adverse cardiac effects. In order to protect clinical trial participants and patients, the International Conference on Harmonization published guidelines to recommend that all new drugs to be tested preclinically for hERG (Kv11.1) channel sensitivity before submitting for regulatory reviews. However, extensive studies have demonstrated that measurement of hERG activity has limitations due to the multiple molecular targets of drug compound through which it may mitigate or abolish a potential arrhythmia, and therefore, a model measuring multiple ion channel effects is likely to be more predictive. Several phenotypic rapid-throughput methods have been developed to predict the potential cardiac toxic compounds in the early stages of drug development using embryonic stem cells- or human induced pluripotent stem cell-derived cardiomyocytes. These rapid-throughput methods include microelectrode array-based field potential assay, impedance-based or Ca(2+) dynamics-based cardiomyocytes contractility assays. This review aims to discuss advantages and limitations of these phenotypic assays for cardiac toxicity assessment.
Collapse
|
28
|
Yan M, Zhang K, Shi Y, Feng L, Lv L, Li B. Mechanism and pharmacological rescue of berberine-induced hERG channel deficiency. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5737-47. [PMID: 26543354 PMCID: PMC4622489 DOI: 10.2147/dddt.s91561] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Berberine (BBR), an isoquinoline alkaloid mainly isolated from plants of Berberidaceae family, is extensively used to treat gastrointestinal infections in clinics. It has been reported that BBR can block human ether-a-go-go-related gene (hERG) potassium channel and inhibit its membrane expression. The hERG channel plays crucial role in cardiac repolarization and is the target of diverse proarrhythmic drugs. Dysfunction of hERG channel can cause long QT syndrome. However, the regulatory mechanisms of BBR effects on hERG at cell membrane level remain unknown. This study was designed to investigate in detail how BBR decreased hERG expression on cell surface and further explore its pharmacological rescue strategies. In this study, BBR decreases caveolin-1 expression in a concentration-dependent manner in human embryonic kidney 293 (HEK293) cells stably expressing hERG channel. Knocking down the basal expression of caveolin-1 alleviates BBR-induced hERG reduction. In addition, we found that aromatic tyrosine (Tyr652) and phenylalanine (Phe656) in S6 domain mediate the long-term effect of BBR on hERG by using mutation techniques. Considering both our previous and present work, we propose that BBR reduces hERG membrane stability with multiple mechanisms. Furthermore, we found that fexofenadine and resveratrol shorten action potential duration prolongated by BBR, thus having the potential effects of alleviating the cardiotoxicity of BBR.
Collapse
Affiliation(s)
- Meng Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Kaiping Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Yanhui Shi
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Lifang Feng
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Lin Lv
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Baoxin Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China ; State-Province Key Laboratory of Biopharmaceutical Engineering, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
29
|
|
30
|
Computational investigations of hERG channel blockers: New insights and current predictive models. Adv Drug Deliv Rev 2015; 86:72-82. [PMID: 25770776 DOI: 10.1016/j.addr.2015.03.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/13/2015] [Accepted: 03/04/2015] [Indexed: 01/08/2023]
Abstract
Identification of potential human Ether-a-go-go Related-Gene (hERG) potassium channel blockers is an essential part of the drug development and drug safety process in pharmaceutical industries or academic drug discovery centers, as they may lead to drug-induced QT prolongation, arrhythmia and Torsade de Pointes. Recent reports also suggest starting to address such issues at the hit selection stage. In order to prioritize molecules during the early drug discovery phase and to reduce the risk of drug attrition due to cardiotoxicity during pre-clinical and clinical stages, computational approaches have been developed to predict the potential hERG blockage of new drug candidates. In this review, we will describe the current in silico methods developed and applied to predict and to understand the mechanism of actions of hERG blockers, including ligand-based and structure-based approaches. We then discuss ongoing research on other ion channels and hERG polymorphism susceptible to be involved in LQTS and how systemic approaches can help in the drug safety decision.
Collapse
|
31
|
Abstract
With the adoption of the ICH E14 guidance, the thorough QT/QTc (TQT) study has become the focus of clinical assessment of an NCE's effects on ECG parameters. The TQT study is used as a guide to the liability of a drug to cause proarrhythmias on the basis of delayed cardiac repolarization. Around 300 TQT studies have been performed since 2005 and through interactions between sponsors and regulators, especially FDA's Interdisciplinary Review Team (IRT) for QT studies. These studies can today be performed more effectively and with great confidence in the generated data. This chapter will discuss technical features and the design and analysis of TQT studies, how assay sensitivity is demonstrated, and examples from recently conducted studies. ECG assessment for drugs that cannot be safely given to healthy volunteers is also addressed, and examples from studies in cancer patients and in healthy volunteers with tyrosine kinase inhibitors are discussed. The TQT study is resource intensive and designed to solely evaluate whether an NCE prolongs the QTc interval. If data with similar confidence can be generated from other studies that are routinely performed as part of the clinical development, this would represent a more optimal use of human resources. Methods and approaches to increase the confidence in ECG data derived from "early QT assessment" in single-ascending/multiple-ascending dose studies are therefore discussed, and a path toward replacing the TQT study using these approaches will be outlined.
Collapse
Affiliation(s)
- Borje Darpo
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd's Hospital, Stockholm, Sweden,
| |
Collapse
|
32
|
Zhang KP, Yang BF, Li BX. Translational toxicology and rescue strategies of the hERG channel dysfunction: biochemical and molecular mechanistic aspects. Acta Pharmacol Sin 2014; 35:1473-84. [PMID: 25418379 PMCID: PMC4261120 DOI: 10.1038/aps.2014.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/20/2014] [Indexed: 01/08/2023]
Abstract
The human ether-à-go-go related gene (hERG) potassium channel is an obligatory anti-target for drug development on account of its essential role in cardiac repolarization and its close association with arrhythmia. Diverse drugs have been removed from the market owing to their inhibitory activity on the hERG channel and their contribution to acquired long QT syndrome (LQTS). Moreover, mutations that cause hERG channel dysfunction may induce congenital LQTS. Recently, an increasing number of biochemical and molecular mechanisms underlying hERG-associated LQTS have been reported. In fact, numerous potential biochemical and molecular rescue strategies are hidden within the biogenesis and regulating network. So far, rescue strategies of hERG channel dysfunction and LQTS mainly include activators, blockers, and molecules that interfere with specific links and other mechanisms. The aim of this review is to discuss the rescue strategies based on hERG channel toxicology from the biochemical and molecular perspectives.
Collapse
Affiliation(s)
- Kai-ping Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| | - Bao-feng Yang
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| | - Bao-xin Li
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| |
Collapse
|
33
|
QTc interval prolongation with vascular endothelial growth factor receptor tyrosine kinase inhibitors. Br J Cancer 2014; 112:296-305. [PMID: 25349964 PMCID: PMC4453446 DOI: 10.1038/bjc.2014.564] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/15/2014] [Accepted: 10/08/2014] [Indexed: 11/08/2022] Open
Abstract
Background: Multi-targeted vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) are known to cause cardiac toxicity, but the relative risk (RR) of QTc interval prolongation and serious arrhythmias associated with them are not reported. Methods: We conducted a trial-level meta-analysis of randomised phase II and III trials comparing arms with and without a US Food and Drug Administration-approved VEGFR TKI (sunitinib, sorafenib, pazopanib, axitinib, vandetanib, cabozantinib, ponatinib and regorafenib). A total of 6548 patients from 18 trials were selected. Statistical analyses were conducted to calculate the summary incidence, RR and 95% CIs. Results: The RR for all-grade and high-grade QTc prolongation for the TKI vs no TKI arms was 8.66 (95% CI 4.92–15.2, P<0.001) and 2.69 (95% CI 1.33–5.44, P=0.006), respectively, with most of the events being asymptomatic QTc prolongation. Respectively, 4.4% and 0.83% of patients exposed to VEGFR TKI had all-grade and high-grade QTc prolongation. On subgroup analysis, only sunitinib and vandetanib were associated with a statistically significant risk of QTc prolongation, with higher doses of vandetanib associated with a greater risk. The rate of serious arrhythmias including torsades de pointes did not seem to be higher with high-grade QTc prolongation. The risk of QTc prolongation was independent of the duration of therapy. Conclusions: In the largest study to date, we show that VEGFR TKI can be associated with QTc prolongation. Although most cases were of low clinical significance, it is unclear whether the same applies to patients treated off clinical trials.
Collapse
|
34
|
Nogawa H, Kawai T. hERG trafficking inhibition in drug-induced lethal cardiac arrhythmia. Eur J Pharmacol 2014; 741:336-9. [DOI: 10.1016/j.ejphar.2014.06.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/16/2014] [Accepted: 06/23/2014] [Indexed: 02/01/2023]
|
35
|
Varkevisser R, Houtman MJC, Linder T, de Git KCG, Beekman HDM, Tidwell RR, Ijzerman AP, Stary-Weinzinger A, Vos MA, van der Heyden MAG. Structure-activity relationships of pentamidine-affected ion channel trafficking and dofetilide mediated rescue. Br J Pharmacol 2014; 169:1322-34. [PMID: 23586323 DOI: 10.1111/bph.12208] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 02/13/2013] [Accepted: 04/04/2013] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Drug interference with normal hERG protein trafficking substantially reduces the channel density in the plasma membrane and thereby poses an arrhythmic threat. The chemical substructures important for hERG trafficking inhibition were investigated using pentamidine as a model drug. Furthermore, the relationship between acute ion channel block and correction of trafficking by dofetilide was studied. EXPERIMENTAL APPROACH hERG and K(IR)2.1 trafficking in HEK293 cells was evaluated by Western blot and immunofluorescence microscopy after treatment with pentamidine and six pentamidine analogues, and correction with dofetilide and four dofetilide analogues that displayed different abilities to inhibit IKr . Molecular dynamics simulations were used to address mode, number and type of interactions between hERG and dofetilide analogues. KEY RESULTS Structural modifications of pentamidine differentially affected plasma membrane levels of hERG and K(IR)2.1. Modification of the phenyl ring or substituents directly attached to it had the largest effect, affirming the importance of these chemical residues in ion channel binding. PA-4 had the mildest effects on both ion channels. Dofetilide corrected pentamidine-induced hERG, but not K(IR)2.1 trafficking defects. Dofetilide analogues that displayed high channel affinity, mediated by pi-pi stacks and hydrophobic interactions, also restored hERG protein levels, whereas analogues with low affinity were ineffective. CONCLUSIONS AND IMPLICATIONS Drug-induced trafficking defects can be minimized if certain chemical features are avoided or 'synthesized out'; this could influence the design and development of future drugs. Further analysis of such features in hERG trafficking correctors may facilitate the design of a non-blocking corrector for trafficking defective hERG proteins in both congenital and acquired LQTS.
Collapse
Affiliation(s)
- R Varkevisser
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Darpo B, Garnett C, Benson CT, Keirns J, Leishman D, Malik M, Mehrotra N, Prasad K, Riley S, Rodriguez I, Sager P, Sarapa N, Wallis R. Cardiac Safety Research Consortium: can the thorough QT/QTc study be replaced by early QT assessment in routine clinical pharmacology studies? Scientific update and a research proposal for a path forward. Am Heart J 2014; 168:262-72. [PMID: 25173536 DOI: 10.1016/j.ahj.2014.06.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 06/03/2014] [Indexed: 11/29/2022]
Abstract
The International Conference on Harmonization E14 guidance for the clinical evaluation of QT/QTc interval prolongation requires almost all new drugs to undergo a dedicated clinical study, primarily in healthy volunteers, the so-called TQT study. Since 2005, when the E14 guidance was implemented in United States and Europe, close to 400 TQT studies have been conducted. In February 2012, the Cardiac Safety Research Consortium held a think tank meeting at Food and Drug Administration's White Oak campus to discuss whether "QT assessment" can be performed as part of routine phase 1 studies. Based on these discussions, a group of experts convened to discuss how to improve the confidence in QT data from early clinical studies, for example, the First-Time-in-Human trial, through collection of serial electrocardiograms and pharmacokinetic samples and the use of exposure response analysis. Recommendations are given on how to design such "early electrocardiogram assessment," and the limitation of not having a pharmacologic-positive control in these studies is discussed. A research path is identified toward collecting evidence to replace or provide an alternative to the dedicated TQT study.
Collapse
Affiliation(s)
- Borje Darpo
- Karolinska Institutet, Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd's Hospital, Stockholm, Sweden; iCardiac Technologies, Rochester, NY.
| | | | | | - James Keirns
- Global Clinical Pharmacology & Exploratory Development, Astellas Pharma Global, Development, Inc, Northbrook, IL
| | - Derek Leishman
- Global PK/PD & Pharmacometrics, Lilly Research Laboratories, Eli Lilly & Co, Indianapolis, IN
| | - Marek Malik
- St Paul's Cardiac Electrophysiology, University of London, and Imperial College, London, United Kingdom
| | - Nitin Mehrotra
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD
| | - Krishna Prasad
- Medicines and Healthcare products Regulatory Agency, DoH, London, United Kingdom
| | - Steve Riley
- Clinical Pharmacology, Global Innovative Pharma Business, Pfizer Inc, Groton, CT
| | - Ignacio Rodriguez
- Pharma Development Safety Risk Management, Roche TCRC, Inc, New York, NY
| | | | - Nenad Sarapa
- Clinical Sciences, Bayer Healthcare, Inc. Whippany, NJ
| | - Robert Wallis
- Safety Pharmacology Consultant, Sandwich, United Kingdom
| |
Collapse
|
37
|
Jewell RC, Laubscher K, Lewis E, Fang L, Gafoor Z, Carey J, McKeown A, West S, Wright O, Sedoti D, Dixon I, Hottenstein CS, Chan G. Assessment of the effect of ofatumumab on cardiac repolarization. J Clin Pharmacol 2014; 55:114-21. [PMID: 25103870 DOI: 10.1002/jcph.376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/04/2014] [Indexed: 11/09/2022]
Abstract
Ofatumumab is a human monoclonal antibody that binds to a unique CD20 epitope on the surface of B lymphocytes, resulting in efficient lysis of CD20-expressing cells via complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity. The potential effect of ofatumumab on cardiac repolarization and the relationship between ofatumumab concentration and change in corrected QT interval (ΔQTcF) were evaluated in data from three clinical trials in 82 patients with chronic lymphocytic leukemia receiving ofatumumab alone (n = 14), ofatumumab with chemotherapy (n = 33), and chemotherapy alone (n = 35). Because of ofatumumab accumulation, baseline QTcF interval was recorded prior to the first infusion for each patient. No patient had a post-baseline QTcF interval >480 milliseconds or a ΔQTcF >60 milliseconds; five patients (four on ofatumumab) had a ΔQTcF between 30 and 60 milliseconds. At cycle 6 (week 21; 308 μg/mL), there was an increase in QTcF in patients on ofatumumab treatment, with an estimated between-treatment difference (90% CI) of 12.5 (4.5, 20.5) milliseconds. However, at the visit with the highest median concentration (week 8; 1386 μg/mL), median ΔQTcF was 4.8 milliseconds. There was no significant relationship between ofatumumab plasma concentration and ΔQTcF. Ofatumumab did not have a clinically significant effect on cardiac repolarization.
Collapse
|
38
|
Heijman J, Voigt N, Carlsson LG, Dobrev D. Cardiac safety assays. Curr Opin Pharmacol 2014; 15:16-21. [DOI: 10.1016/j.coph.2013.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/04/2013] [Accepted: 11/07/2013] [Indexed: 12/22/2022]
|
39
|
Pharmacological chaperoning: a primer on mechanism and pharmacology. Pharmacol Res 2014; 83:10-9. [PMID: 24530489 DOI: 10.1016/j.phrs.2014.01.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 12/26/2022]
Abstract
Approximately forty percent of diseases are attributable to protein misfolding, including those for which genetic mutation produces misfolding mutants. Intriguingly, many of these mutants are not terminally misfolded since native-like folding, and subsequent trafficking to functional locations, can be induced by target-specific, small molecules variably termed pharmacological chaperones, pharmacoperones, or pharmacochaperones (PCs). PC targets include enzymes, receptors, transporters, and ion channels, revealing the breadth of proteins that can be engaged by ligand-assisted folding. The purpose of this review is to provide an integrated primer of the diverse mechanisms and pharmacology of PCs. In this regard, we examine the structural mechanisms that underlie PC rescue of misfolding mutants, including the ability of PCs to act as surrogates for defective intramolecular interactions and, at the intermolecular level, overcome oligomerization deficiencies and dominant negative effects, as well as influence the subunit stoichiometry of heteropentameric receptors. Not surprisingly, PC-mediated structural correction of misfolding mutants normalizes interactions with molecular chaperones that participate in protein quality control and forward-trafficking. A variety of small molecules have proven to be efficacious PCs and the advantages and disadvantages of employing orthostatic antagonists, active-site inhibitors, orthostatic agonists, and allosteric modulator PCs are considered. Also examined is the possibility that several therapeutic agents may have unrecognized activity as PCs, and this chaperoning activity may mediate/contribute to therapeutic action and/or account for adverse effects. Lastly, we explore evidence that pharmacological chaperoning exploits intrinsic ligand-assisted folding mechanisms. Given the widespread applicability of PC rescue of mutants associated with protein folding disorders, both in vitro and in vivo, the therapeutic potential of PCs is vast. This is most evident in the treatment of lysosomal storage disorders, cystic fibrosis, and nephrogenic diabetes insipidus, for which proof of principle in humans has been demonstrated.
Collapse
|
40
|
Townsend C, Brown BS. Predicting drug-induced QT prolongation and torsades de pointes: a review of preclinical endpoint measures. ACTA ACUST UNITED AC 2013; Chapter 10:Unit 10.16. [PMID: 23744708 DOI: 10.1002/0471141755.ph1016s61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Compound-induced prolongation of the cardiac QT interval is a major concern in drug development and this unit discusses approaches that can predict QT effects prior to undertaking clinical trials. The majority of compounds that prolong the QT interval block the cardiac rapid delayed rectifier potassium current, IKr (hERG). Described in this overview are different ways to measure hERG, from recent advances in automated electrophysiology to the quantification of channel protein trafficking and binding. The contribution of other cardiac ion channels to hERG data interpretation is also discussed. In addition, endpoint measures of the integrated activity of cardiac ion channels at the single-cell, tissue, and whole-animal level, including for example the well-established action potential to the more recent beat-to-beat variability, transmural dispersion of repolarization, and field potential duration, are described in the context of their ability to predict QT prolongation and torsadogenicity in humans.
Collapse
Affiliation(s)
- Claire Townsend
- GlaxoSmithKline Biological Reagents and Assay Development, Research Triangle Park, NC, USA
| | | |
Collapse
|
41
|
Apaja PM, Foo B, Okiyoneda T, Valinsky WC, Barriere H, Atanasiu R, Ficker E, Lukacs GL, Shrier A. Ubiquitination-dependent quality control of hERG K+ channel with acquired and inherited conformational defect at the plasma membrane. Mol Biol Cell 2013; 24:3787-804. [PMID: 24152733 PMCID: PMC3861077 DOI: 10.1091/mbc.e13-07-0417] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Membrane trafficking in concert with the peripheral quality control machinery plays a critical role in preserving plasma membrane (PM) protein homeostasis. Unfortunately, the peripheral quality control may also dispose of partially or transiently unfolded polypeptides and thereby contribute to the loss-of-expression phenotype of conformational diseases. Defective functional PM expression of the human ether-a-go-go-related gene (hERG) K(+) channel leads to the prolongation of the ventricular action potential that causes long QT syndrome 2 (LQT2), with increased propensity for arrhythmia and sudden cardiac arrest. LQT2 syndrome is attributed to channel biosynthetic processing defects due to mutation, drug-induced misfolding, or direct channel blockade. Here we provide evidence that a peripheral quality control mechanism can contribute to development of the LQT2 syndrome. We show that PM hERG structural and metabolic stability is compromised by the reduction of extracellular or intracellular K(+) concentration. Cardiac glycoside-induced intracellular K(+) depletion conformationally impairs the complex-glycosylated channel, which provokes chaperone- and C-terminal Hsp70-interacting protein-dependent polyubiquitination, accelerated internalization, and endosomal sorting complex required for transport-dependent lysosomal degradation. A similar mechanism contributes to the down-regulation of PM hERG harboring LQT2 missense mutations, with incomplete secretion defect. These results suggest that PM quality control plays a determining role in the loss-of-expression phenotype of hERG in certain hereditary and acquired LTQ2 syndromes.
Collapse
Affiliation(s)
- Pirjo M Apaja
- Department of Physiology and Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montréal, QC H3E 1Y6, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
A randomized, double-blind, placebo-controlled study to assess QTc interval prolongation of standard dose aflibercept in cancer patients treated with docetaxel. J Cardiovasc Pharmacol 2013; 61:495-504. [PMID: 23429593 DOI: 10.1097/fjc.0b013e31828b73ff] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
: The effect of repeated doses of aflibercept on ventricular repolarization in cancer patients was evaluated in an intensive electrocardiogram trial. This randomized, placebo-controlled, double-blind trial was conducted in 87 treated solid tumor patients. Treatment was with 6 mg/kg aflibercept, 1-hour intravenous (n = 43), or placebo (n = 44), combined with ≤75 mg/m docetaxel, every 3 weeks. Electrocardiograms were collected for 6 hours posttreatment using digital 12-lead Holter recorders, at day 1, in cycles 1 and 3. Free and vascular endothelial growth factor-bound aflibercept concentrations were assessed at similar time points. Eighty-four patients (43 placebo and 41 aflibercept) were evaluable for QT interval, Fridericia correction (QTcF) at cycle 1 and 59 (31 placebo and 28 aflibercept) at cycle 3. During cycle 3, from 30 minutes to 6 hours after the start of aflibercept, the maximum observed upper limit of the QTcF 90% confidence interval was 16 ms, for a mean of 8.4 ms. QTcF prolongation above 480 ms and 60 ms above baseline was observed in 1 aflibercept patient (2%). The slope of the relationship between free aflibercept concentration and QTcF was 0.048 (95% confidence interval, 0.013-0.082), corresponding to a 5-ms increase per 100 µg/mL increase in concentration. These results exclude a clinically important effect of aflibercept on ventricular repolarization.
Collapse
|
43
|
Cardiac ion channel trafficking defects and drugs. Pharmacol Ther 2013; 139:24-31. [DOI: 10.1016/j.pharmthera.2013.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 03/14/2013] [Indexed: 01/19/2023]
|
44
|
Rampe D, Brown AM. A history of the role of the hERG channel in cardiac risk assessment. J Pharmacol Toxicol Methods 2013; 68:13-22. [DOI: 10.1016/j.vascn.2013.03.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/14/2013] [Accepted: 03/14/2013] [Indexed: 01/25/2023]
|
45
|
Himmel HM. Drug-induced functional cardiotoxicity screening in stem cell-derived human and mouse cardiomyocytes: effects of reference compounds. J Pharmacol Toxicol Methods 2013; 68:97-111. [PMID: 23702537 DOI: 10.1016/j.vascn.2013.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/10/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Early prediction of drug-induced functional cardiotoxicity requires robust in-vitro systems suitable for medium/high throughput and easily accessible cardiomyocytes with defined reproducible properties. The xCELLigence Cardio system uses 96-well plates with interdigitated electrodes that detect the impedance changes of rhythmic contractions of stem cell-derived cardiomyocyte (SC-CM) layers. Here, we report on our initial screening experience in comparison to established (multi)cellular and in-vivo models. METHODS Impedance signals from human iPSC-CM (iCells™) and mouse eSC-CM (Cor.At™) were analyzed for contraction amplitude (CA) and duration, rise/fall time, beating rate (BR) and irregularity. RESULTS Following solution exchange, impedance signals re-approximated steady-state conditions after about 2 (Cor.At™) and 3h (iCells™); these time points were used to analyze drug effects. The solvent DMSO (≤1%) hardly influenced contraction parameters in Cor.At™, whereas in iCells™ DMSO (>0.1%) reduced CA and enhanced BR. The selective hERG K⁺ channel blockers E-4031 and dofetilide reduced CA and accelerated BR (≥30 nM) according to the analysis software. The latter, however, was due to burst-like contractions (300 nM) that could be detected only by visual inspection of recordings, and were more pronounced in Cor.At™ as in iCells™. In cardiac myocytes and tissue preparations, however, E4031 and dofetilide have been reported to increase cell shortening and contractile force and to reduce BR. Compounds (pentamidine, HMR1556, ATX2, TTX, and verapamil) with other mechanisms of action were also investigated; their effects differed partially between cell lines (e.g. TTX) and compared to established (multi)cellular models (e.g. HMR1556, ouabain). CONCLUSION Mouse and human stem cell-derived cardiomyocytes respond differently to drugs and these responses occasionally also differ from those originating from established in-vitro and in-vivo models. Hence, drug-induced cardiotoxic effects may be detected with this system, however, the predictive or even translational value of results is considered limited and not yet firmly established.
Collapse
|
46
|
Takanari H, Nalos L, Stary-Weinzinger A, de Git KCG, Varkevisser R, Linder T, Houtman MJC, Peschar M, de Boer TP, Tidwell RR, Rook MB, Vos MA, van der Heyden MAG. Efficient and specific cardiac IK1 inhibition by a new pentamidine analogue. Cardiovasc Res 2013; 99:203-14. [DOI: 10.1093/cvr/cvt103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
47
|
Inhibiting the clathrin-mediated endocytosis pathway rescues KIR2.1 downregulation by pentamidine. Pflugers Arch 2012. [DOI: 10.1007/s00424-012-1189-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
48
|
Zhang Y, Dong Z, Jin L, Zhang K, Zhao X, Fu J, Gong Y, Sun M, Yang B, Li B. Arsenic trioxide-induced hERG K(+) channel deficiency can be rescued by matrine and oxymatrine through up-regulating transcription factor Sp1 expression. Biochem Pharmacol 2012; 85:59-68. [PMID: 23103450 DOI: 10.1016/j.bcp.2012.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Revised: 09/02/2012] [Accepted: 09/04/2012] [Indexed: 11/28/2022]
Abstract
The human ether-a-go-go-related gene (hERG) encodes the rapidly activating, delayed rectifier potassium channel (IKr) important for cardiac repolarization. Dysfunction of the hERG channel can cause Long QT Syndrome (LQTS). A wide variety of structurally diverse therapeutic compounds reduce the hERG current by acute direct inhibition of the hERG current or/and selective disruption of hERG protein expression. 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. This cardiotoxicity limits its clinical applications. Our aim was to develop cardioprotective agents to decrease As(2)O(3)-induced cardiotoxicity. We reported that superfusion of hERG-expressing HEK293 (hERG-HEK) cells with matrine (1, 10 μM) increased the hERG current by promoting hERG channel activation. Long-term treatment with 1 μM matrine or oxymatrine increased expression of the hERG protein and rescued the hERG surface expression disrupted by As(2)O(3). In addition, Matrine and oxymatrine significantly shortened action potential duration prolonged by As(2)O(3) in guinea pig ventricular myocytes. These results were ascribed to the up-regulation of hERG at both mRNA and protein levels via an increase in the expression of transcription factor Sp1, an established transactivator of the hERG gene. 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.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, 150086, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jonsson MKB, van der Heyden MAG, van Veen TAB. Deciphering hERG channels: molecular basis of the rapid component of the delayed rectifier potassium current. J Mol Cell Cardiol 2012; 53:369-74. [PMID: 22742967 DOI: 10.1016/j.yjmcc.2012.06.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 06/07/2012] [Accepted: 06/19/2012] [Indexed: 12/23/2022]
Abstract
The rapid component of the delayed rectifier potassium current (I(Kr)), encoded by the ether-a-go-go-related gene (ERG1, officially denominated as KCNH2), is a major contributor to repolarization in the mammalian heart. Acute (e.g. drug-induced) and chronic (e.g. inherited genetic disorder) disruptions of this current can lead to prolongation of the action potential and potentiate occurrence of lethal arrhythmias. Many cardiac and non-cardiac drugs show high affinity for the I(Kr) channel and it is therefore extensively studied during safety pharmacology. The unique biophysical and pharmacological properties of the I(Kr) channel are largely recapitulated by expressing the human variant (hERG1a) in overexpressing systems. hERG1a channels are tetramers consisting of four 1159 amino acid long proteins and have electrophysiological properties similar, but not identical, to native I(Kr). In the search for an explanation to the discrepancies between I(Kr) and hERG1a channels, two alternative hERG1 proteins have been found. Alternative transcription of hERG1 leads to a protein with a 56 amino acid shorter N-terminus, known as hERG1b. hERG1b can form channels alone or coassemble with hERG1a. Alternative splicing leads to an alternate C-terminus and a protein known as hERGuso. hERGuso and hERG1b regulate hERG1a channel trafficking, functional expression and channel kinetics. Expression of hERGuso leads to a reduced number of channels at the plasma membrane and thereby reduces current density. On the contrary, co-assembly with hERG1b alters channel kinetics resulting in more available channels and a larger current. These findings have implication for understanding mechanisms of disease, acute and chronic drug effects, and potential gender differences.
Collapse
Affiliation(s)
- Malin K B Jonsson
- Department of Medical Physiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|