1
|
Li E, van der Heyden MAG. The network of cardiac K IR2.1: its function, cellular regulation, electrical signaling, diseases and new drug avenues. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6369-6389. [PMID: 38683369 PMCID: PMC11422472 DOI: 10.1007/s00210-024-03116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
The functioning of the human heart relies on complex electrical and communication systems that coordinate cardiac contractions and sustain rhythmicity. One of the key players contributing to this intricate system is the KIR2.1 potassium ion channel, which is encoded by the KCNJ2 gene. KIR2.1 channels exhibit abundant expression in both ventricular myocytes and Purkinje fibers, exerting an important role in maintaining the balance of intracellular potassium ion levels within the heart. And by stabilizing the resting membrane potential and contributing to action potential repolarization, these channels have an important role in cardiac excitability also. Either gain- or loss-of-function mutations, but also acquired impairments of their function, are implicated in the pathogenesis of diverse types of cardiac arrhythmias. In this review, we aim to elucidate the system functions of KIR2.1 channels related to cellular electrical signaling, communication, and their contributions to cardiovascular disease. Based on this knowledge, we will discuss existing and new pharmacological avenues to modulate their function.
Collapse
Affiliation(s)
- Encan Li
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM, Utrecht, Netherlands
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM, Utrecht, Netherlands.
| |
Collapse
|
2
|
Li E, Kool W, Woolschot L, van der Heyden MAG. Chronic Propafenone Application Increases Functional K IR2.1 Expression In Vitro. Pharmaceuticals (Basel) 2023; 16:ph16030404. [PMID: 36986503 PMCID: PMC10056987 DOI: 10.3390/ph16030404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Expression and activity of inwardly rectifying potassium (KIR) channels within the heart are strictly regulated. KIR channels have an important role in shaping cardiac action potentials, having a limited conductance at depolarized potentials but contributing to the final stage of repolarization and resting membrane stability. Impaired KIR2.1 function causes Andersen-Tawil Syndrome (ATS) and is associated with heart failure. Restoring KIR2.1 function by agonists of KIR2.1 (AgoKirs) would be beneficial. The class 1c antiarrhythmic drug propafenone is identified as an AgoKir; however, its long-term effects on KIR2.1 protein expression, subcellular localization, and function are unknown. Propafenone's long-term effect on KIR2.1 expression and its underlying mechanisms in vitro were investigated. KIR2.1-carried currents were measured by single-cell patch-clamp electrophysiology. KIR2.1 protein expression levels were determined by Western blot analysis, whereas conventional immunofluorescence and advanced live-imaging microscopy were used to assess the subcellular localization of KIR2.1 proteins. Acute propafenone treatment at low concentrations supports the ability of propafenone to function as an AgoKir without disturbing KIR2.1 protein handling. Chronic propafenone treatment (at 25-100 times higher concentrations than in the acute treatment) increases KIR2.1 protein expression and KIR2.1 current densities in vitro, which are potentially associated with pre-lysosomal trafficking inhibition.
Collapse
Affiliation(s)
- Encan Li
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Willy Kool
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Liset Woolschot
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
3
|
Xynogalos P, Rahm AK, Fried S, Chasan S, Scherer D, Seyler C, Katus HA, Frey N, Zitron E. Verapamil inhibits Kir2.3 channels by binding to the pore and interfering with PIP2 binding. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:659-667. [PMID: 36445385 PMCID: PMC10042922 DOI: 10.1007/s00210-022-02342-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
Abstract
Abstract
The inwardly rectifying potassium current of the cardiomyocyte (IK1) is the main determinant of the resting potential. Ion channels Kir2.1, Kir2.2, and Kir2.3 form tetramers and are the molecular correlate of macroscopic IK1 current. Verapamil is an antiarrhythmic drug used to suppress atrial and ventricular arrhythmias. Its primary mechanism of action is via blocking calcium channels. In addition, it has been demonstrated to block IK1 current and the Kir2.1 subunit. Its effect on other subunits that contribute to IK1 current has not been studied to date. We therefore analyzed the effect of verapamil on the Kir channels 2.1, 2.2, and 2.3 in the Xenopus oocyte expression system. Kir2.1, Kir2.2, and Kir2.3 channels were heterologously expressed in Xenopus oocytes. Respective currents were measured with the voltage clamp technique and the effect of verapamil on the current was measured. At a concentration of 300 µM, verapamil inhibited Kir2.1 channels by 41.36% ± 2.7 of the initial current, Kir2.2 channels by 16.51 ± 3.6%, and Kir2.3 by 69.98 ± 4.2%. As a verapamil effect on kir2.3 was a previously unknown finding, we analyzed this effect further. At wash in with 300 µM verapamil, the maximal effect was seen within 20 min of the infusion. After washing out with control solution, there was only a partial current recovery. The current reduction from verapamil was the same at − 120 mV (73.2 ± 3.7%), − 40 mV (85.5 ± 6.5%), and 0 mV (61.5 ± 10.6%) implying no voltage dependency of the block. Using site directed mutations in putative binding sites, we demonstrated a decrease of effect with pore mutant E291A and absence of verapamil effect for D251A. With mutant I214L, which shows a stronger affinity for PIP2 binding, we observed a normalized current reduction to 61.9 ± 0.06% of the control current, which was significantly less pronounced compared to wild type channels. Verapamil blocks Kir2.1, Kir2.2, and Kir2.3 subunits. In Kir2.3, blockade is dependent on sites E291 and D251 and interferes with activation of the channel via PIP2. Interference with these sites and with PIP2 binding has also been described for other Kir channels blocking drugs. As Kir2.3 is preferentially expressed in atrium, a selective Kir2.3 blocking agent would constitute an interesting antiarrhythmic concept.
Collapse
|
4
|
Houtman MJC, Friesacher T, Chen X, Zangerl-Plessl EM, van der Heyden MAG, Stary-Weinzinger A. Development of I KATP Ion Channel Blockers Targeting Sulfonylurea Resistant Mutant K IR6.2 Based Channels for Treating DEND Syndrome. Front Pharmacol 2022; 12:814066. [PMID: 35095528 PMCID: PMC8795863 DOI: 10.3389/fphar.2021.814066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: DEND syndrome is a rare channelopathy characterized by a combination of developmental delay, epilepsy and severe neonatal diabetes. Gain of function mutations in the KCNJ11 gene, encoding the KIR6.2 subunit of the IKATP potassium channel, stand at the basis of most forms of DEND syndrome. In a previous search for existing drugs with the potential of targeting Cantú Syndrome, also resulting from increased IKATP, we found a set of candidate drugs that may also possess the potential to target DEND syndrome. In the current work, we combined Molecular Modelling including Molecular Dynamics simulations, with single cell patch clamp electrophysiology, in order to test the effect of selected drug candidates on the KIR6.2 WT and DEND mutant channels. Methods: Molecular dynamics simulations were performed to investigate potential drug binding sites. To conduct in vitro studies, KIR6.2 Q52R and L164P mutants were constructed. Inside/out patch clamp electrophysiology on transiently transfected HEK293T cells was performed for establishing drug-channel inhibition relationships. Results: Molecular Dynamics simulations provided insight in potential channel interaction and shed light on possible mechanisms of action of the tested drug candidates. Effective IKIR6.2/SUR2a inhibition was obtained with the pore-blocker betaxolol (IC50 values 27-37 μM). Levobetaxolol effectively inhibited WT and L164P (IC50 values 22 μM) and Q52R (IC50 55 μM) channels. Of the SUR binding prostaglandin series, travoprost was found to be the best blocker of WT and L164P channels (IC50 2-3 μM), while Q52R inhibition was 15-20% at 10 μM. Conclusion: Our combination of MD and inside-out electrophysiology provides the rationale for drug mediated IKATP inhibition, and will be the basis for 1) screening of additional existing drugs for repurposing to address DEND syndrome, and 2) rationalized medicinal chemistry to improve IKATP inhibitor efficacy and specificity.
Collapse
Affiliation(s)
- Marien J C Houtman
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Theres Friesacher
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Xingyu Chen
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Eva-Maria Zangerl-Plessl
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Anna Stary-Weinzinger
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Sanderson L, da Silva M, Sekhar GN, Brown RC, Burrell-Saward H, Fidanboylu M, Liu B, Dailey LA, Dreiss CA, Lorenz C, Christie M, Persaud SJ, Yardley V, Croft SL, Valero M, Thomas SA. Drug reformulation for a neglected disease. The NANOHAT project to develop a safer more effective sleeping sickness drug. PLoS Negl Trop Dis 2021; 15:e0009276. [PMID: 33857146 PMCID: PMC8078842 DOI: 10.1371/journal.pntd.0009276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2021] [Accepted: 02/26/2021] [Indexed: 01/16/2023] Open
Abstract
Background Human African trypanosomiasis (HAT or sleeping sickness) is caused by the
parasite Trypanosoma brucei sspp. The disease has two
stages, a haemolymphatic stage after the bite of an infected tsetse fly,
followed by a central nervous system stage where the parasite penetrates the
brain, causing death if untreated. Treatment is stage-specific, due to the
blood-brain barrier, with less toxic drugs such as pentamidine used to treat
stage 1. The objective of our research programme was to develop an
intravenous formulation of pentamidine which increases CNS exposure by some
10–100 fold, leading to efficacy against a model of stage 2 HAT. This target
candidate profile is in line with drugs for neglected diseases inititative
recommendations. Methodology To do this, we evaluated the physicochemical and structural characteristics
of formulations of pentamidine with Pluronic micelles (triblock-copolymers
of polyethylene-oxide and polypropylene oxide), selected candidates for
efficacy and toxicity evaluation in vitro, quantified
pentamidine CNS delivery of a sub-set of formulations in vitro and
in vivo, and progressed one pentamidine-Pluronic formulation
for further evaluation using an in vivo single dose brain
penetration study. Principal Findings Screening pentamidine against 40 CNS targets did not reveal any major
neurotoxicity concerns, however, pentamidine had a high affinity for the
imidazoline2 receptor. The reduction in insulin secretion in
MIN6 β-cells by pentamidine may be secondary to pentamidine-mediated
activation of β-cell imidazoline receptors and impairment of cell viability.
Pluronic F68 (0.01%w/v)-pentamidine formulation had a similar inhibitory
effect on insulin secretion as pentamidine alone and an additive
trypanocidal effect in vitro. However, all Pluronics tested
(P85, P105 and F68) did not significantly enhance brain exposure of
pentamidine. Significance These results are relevant to further developing block-copolymers as
nanocarriers, improving BBB drug penetration and understanding the side
effects of pentamidine. Sleeping sickness or human African Trypanosomiasis (HAT) is a disease caused by a
parasite, which is transferred to humans by the bite of an infected tsetse fly.
There are two disease stages: the first stage is the blood-based stage of the
disease and the second stage affects the brain. It is fatal if left untreated.
The blood-brain barrier (BBB) makes the brain stage difficult to treat because
it prevents 99% of all drugs from entering the brain from the blood. Those
anti-HAT drugs that do enter the brain are toxic and have serious side effects.
Pentamidine is a less toxic blood stage drug, which our research has shown has a
limited ability to cross the BBB due to its removal by proteins called
transporters. The objective of this study was to use Pluronic to improve
pentamidine delivery to target sites, whilst reducing its side effects. Pluronic
is a polymer, which can assemble into micelles and encapsulate the drug. Thus,
prolonging its circulation time and protecting it. Our study indicated that the
selected Pluronics did not increase the brain delivery of pentamidine. However.
Pluronic-pentamidine formulations were identified that harboured trypanocidal
activity and did not increase safety concerns compared to unformulated
pentamidine.
Collapse
Affiliation(s)
- Lisa Sanderson
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Marcelo da Silva
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Gayathri N. Sekhar
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Rachel C. Brown
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Hollie Burrell-Saward
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and
Tropical Medicine, London, United Kingdom
| | - Mehmet Fidanboylu
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Bo Liu
- King’s College London, Department of Diabetes, School of Life Course
Sciences, Faculty of Life Sciences & Medicine, London, United
Kingdom
| | - Lea Ann Dailey
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Cécile A. Dreiss
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Chris Lorenz
- King’s College London, Theory & Simulation of Condensed Matter Group,
Department of Physics, Strand, London, United Kingdom
| | - Mark Christie
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
| | - Shanta J. Persaud
- King’s College London, Department of Diabetes, School of Life Course
Sciences, Faculty of Life Sciences & Medicine, London, United
Kingdom
| | - Vanessa Yardley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and
Tropical Medicine, London, United Kingdom
| | - Simon L. Croft
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and
Tropical Medicine, London, United Kingdom
| | - Margarita Valero
- Physical Chemistry Department, Faculty of Pharmacy, University of
Salamanca, Salamanca, Spain
| | - Sarah A. Thomas
- King’s College London, Institute of Pharmaceutical Science,
Franklin-Wilkins Building, Stamford Street, London, United
Kingdom
- * E-mail:
| |
Collapse
|
6
|
Weaver CD, Denton JS. Next-generation inward rectifier potassium channel modulators: discovery and molecular pharmacology. Am J Physiol Cell Physiol 2021; 320:C1125-C1140. [PMID: 33826405 DOI: 10.1152/ajpcell.00548.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inward rectifying potassium (Kir) channels play important roles in both excitable and nonexcitable cells of various organ systems and could represent valuable new drug targets for cardiovascular, metabolic, immune, and neurological diseases. In nonexcitable epithelial cells of the kidney tubule, for example, Kir1.1 (KCNJ1) and Kir4.1 (KCNJ10) are linked to sodium reabsorption in the thick ascending limb of Henle's loop and distal convoluted tubule, respectively, and have been explored as novel-mechanism diuretic targets for managing hypertension and edema. G protein-coupled Kir channels (Kir3) channels expressed in the central nervous system are critical effectors of numerous signal transduction pathways underlying analgesia, addiction, and respiratory-depressive effects of opioids. The historical dearth of pharmacological tool compounds for exploring the therapeutic potential of Kir channels has led to a molecular target-based approach using high-throughput screen (HTS) of small-molecule libraries and medicinal chemistry to develop "next-generation" Kir channel modulators that are both potent and specific for their targets. In this article, we review recent efforts focused specifically on discovery and improvement of target-selective molecular probes. The reader is introduced to fluorescence-based thallium flux assays that have enabled much of this work and then provided with an overview of progress made toward developing modulators of Kir1.1 (VU590, VU591), Kir2.x (ML133), Kir3.X (ML297, GAT1508, GiGA1, VU059331), Kir4.1 (VU0134992), and Kir7.1 (ML418). We discuss what is known about the small molecules' molecular mechanisms of action, in vitro and in vivo pharmacology, and then close with our view of what critical work remains to be done.
Collapse
Affiliation(s)
- C David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - Jerod S Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
7
|
Chastain DB, Veve MP, Wagner JL. Abnormal QTc syndrome in HIV-infected patients: a systematic review of prevalence and risk factors. Antivir Ther 2020; 24:459-465. [PMID: 31570667 DOI: 10.3851/imp3335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND The purpose of this review is to critically analyse data regarding the prevalence and risk factors for developing a prolonged QTc interval and subsequent sudden cardiac death (SCD) in persons living with HIV (PLWH). METHODS A systematic literature search using PubMed and Google Scholar databases was performed using the following search terms: 'HIV and prolonged QTc' and 'managing HIV-patients with prolonged QTc'. References within articles of interest were also evaluated. RESULTS/DISCUSSION PLWH are at an increased risk of having a prolonged QTc interval. Some risk factors for this include the virus itself, concomitant medications, comorbid conditions, addictions and electrolyte disturbances. PLWH who have an increased HIV RNA viral load or decreased CD4+ T-cell count are at further risk for progressing to sudden cardiac death (SCD). Many medications commonly prescribed in the PLWH population, such as antiretrovirals and antimicrobials used in opportunistic infection prophylaxis, have also been shown to promote QTc prolongation through inhibition of human ether-a-go-go potassium channels or through drug metabolism inhibition of other QTc prolonging drugs. CONCLUSIONS Due to the high number of risk factors associated with QTc prolongation, clinicians should incorporate baseline and routine ECG monitoring for PLWH to potentially lower the increased risk of SCD in PLWH.
Collapse
Affiliation(s)
- Daniel B Chastain
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Albany, GA, USA
| | - Michael P Veve
- Department of Clinical Pharmacy and Translational Science, University of Tennessee College of Pharmacy, Knoxville, TN, USA
| | - Jamie L Wagner
- Department of Pharmacy Practice, University of Mississippi School of Pharmacy, Jackson, MS, USA
| |
Collapse
|
8
|
Chen X, Garon A, Wieder M, Houtman MJC, Zangerl-Plessl EM, Langer T, van der Heyden MAG, Stary-Weinzinger A. Computational Identification of Novel Kir6 Channel Inhibitors. Front Pharmacol 2019; 10:549. [PMID: 31178728 PMCID: PMC6543810 DOI: 10.3389/fphar.2019.00549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/01/2019] [Indexed: 12/25/2022] Open
Abstract
KATP channels consist of four Kir6.x pore-forming subunits and four regulatory sulfonylurea receptor (SUR) subunits. These channels couple the metabolic state of the cell to membrane excitability and play a key role in physiological processes such as insulin secretion in the pancreas, protection of cardiac muscle during ischemia and hypoxic vasodilation of arterial smooth muscle cells. Abnormal channel function resulting from inherited gain or loss-of-function mutations in either the Kir6.x and/or SUR subunits are associated with severe diseases such as neonatal diabetes, congenital hyperinsulinism, or Cantú syndrome (CS). CS is an ultra-rare genetic autosomal dominant disorder, caused by dominant gain-of-function mutations in SUR2A or Kir6.1 subunits. No specific pharmacotherapeutic treatment options are currently available for CS. Kir6 specific inhibitors could be beneficial for the development of novel drug therapies for CS, particular for mutations, which lack high affinity for sulfonylurea inhibitor glibenclamide. By applying a combination of computational methods including atomistic MD simulations, free energy calculations and pharmacophore modeling, we identified several novel Kir6.1 inhibitors, which might be possible candidates for drug repurposing. The in silico predictions were confirmed using inside/out patch-clamp analysis. Importantly, Cantú mutation C166S in Kir6.2 (equivalent to C176S in Kir6.1) and S1020P in SUR2A, retained high affinity toward the novel inhibitors. Summarizing, the inhibitors identified in this study might provide a starting point toward developing novel therapies for Cantú disease.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Arthur Garon
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Marcus Wieder
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Marien J. C. Houtman
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Thierry Langer
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Marcel A. G. van der Heyden
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | | |
Collapse
|
9
|
Aréchiga-Figueroa IA, Marmolejo-Murillo LG, Cui M, Delgado-Ramírez M, van der Heyden MAG, Sánchez-Chapula JA, Rodríguez-Menchaca AA. High-potency block of Kir4.1 channels by pentamidine: Molecular basis. Eur J Pharmacol 2017; 815:56-63. [PMID: 28993158 DOI: 10.1016/j.ejphar.2017.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Inward rectifier potassium (Kir) channels are expressed in almost all mammalian tissues and contribute to a wide range of physiological processes. Kir4.1 channel expression is found in the brain, inner ear, eye, and kidney. Loss-of-function mutations in the pore-forming Kir4.1 subunit cause an autosomal recessive disorder characterized by epilepsy, ataxia, sensorineural deafness and tubulopathy (SeSAME/EST syndrome). Despite its importance in physiological and pathological conditions, pharmacological research of Kir4.1 is limited. Here, we characterized the effect of pentamidine on Kir4.1 channels using electrophysiology, mutagenesis and computational methods. Pentamidine potently inhibited Kir4.1 channels when applied to the cytoplasmic side under inside-out patch clamp configuration (IC50 = 97nM). The block was voltage dependent. Molecular modeling predicted the binding of pentamidine to the transmembrane pore region of Kir4.1 at aminoacids T127, T128 and E158. Mutation of each of these residues reduced the potency of pentamidine to block Kir4.1 channels. A pentamidine analog (PA-6) inhibited Kir4.1 with similar potency (IC50 = 132nM). Overall, this study shows that pentamidine blocks Kir4.1 channels interacting with threonine and glutamate residues in the transmembrane pore region. These results can be useful to design novel compounds with major potency and specificity over Kir4.1 channels.
Collapse
Affiliation(s)
- Iván A Aréchiga-Figueroa
- CONACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, Mexico
| | | | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Mayra Delgado-Ramírez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, SLP, Mexico
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - José A Sánchez-Chapula
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Col, Mexico
| | - Aldo A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, SLP, Mexico.
| |
Collapse
|
10
|
Ji Y, Veldhuis MG, Zandvoort J, Romunde FL, Houtman MJC, Duran K, van Haaften G, Zangerl-Plessl EM, Takanari H, Stary-Weinzinger A, van der Heyden MAG. PA-6 inhibits inward rectifier currents carried by V93I and D172N gain-of-function K IR2.1 channels, but increases channel protein expression. J Biomed Sci 2017; 24:44. [PMID: 28711067 PMCID: PMC5513211 DOI: 10.1186/s12929-017-0352-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/11/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The inward rectifier potassium current IK1 contributes to a stable resting membrane potential and phase 3 repolarization of the cardiac action potential. KCNJ2 gain-of-function mutations V93I and D172N associate with increased IK1, short QT syndrome type 3 and congenital atrial fibrillation. Pentamidine-Analogue 6 (PA-6) is an efficient (IC50 = 14 nM with inside-out patch clamp methodology) and specific IK1 inhibitor that interacts with the cytoplasmic pore region of the KIR2.1 ion channel, encoded by KCNJ2. At 10 μM, PA-6 increases wild-type (WT) KIR2.1 expression in HEK293T cells upon chronic treatment. We hypothesized that PA-6 will interact with and inhibit V93I and D172N KIR2.1 channels, whereas impact on channel expression at the plasma membrane requires higher concentrations. METHODS Molecular modelling was performed with the human KIR2.1 closed state homology model using FlexX. WT and mutant KIR2.1 channels were expressed in HEK293 cells. Patch-clamp single cell electrophysiology measurements were performed in the whole cell and inside-out mode of the patch clamp method. KIR2.1 expression level and localization were determined by western blot analysis and immunofluorescence microscopy, respectively. RESULTS PA-6 docking in the V93I/D172N double mutant homology model of KIR2.1 demonstrated that mutations and drug-binding site are >30 Å apart. PA-6 inhibited WT and V93I outward currents with similar potency (IC50 = 35.5 and 43.6 nM at +50 mV for WT and V93I), whereas D172N currents were less sensitive (IC50 = 128.9 nM at +50 mV) using inside-out patch-clamp electrophysiology. In whole cell mode, 1 μM of PA-6 inhibited outward IK1 at -50 mV by 28 ± 36%, 18 ± 20% and 10 ± 6%, for WT, V93I and D172N channels respectively. Western blot analysis demonstrated that PA-6 (5 μM, 24 h) increased KIR2.1 expression levels of WT (6.3 ± 1.5 fold), and V93I (3.9 ± 0.9) and D172N (4.8 ± 2.0) mutants. Immunofluorescent microscopy demonstrated dose-dependent intracellular KIR2.1 accumulation following chronic PA-6 application (24 h, 1 and 5 μM). CONCLUSIONS 1) KCNJ2 gain-of-function mutations V93I and D172N in the KIR2.1 ion channel do not impair PA-6 mediated inhibition of IK1, 2) PA-6 elevates KIR2.1 protein expression and induces intracellular KIR2.1 accumulation, 3) PA-6 is a strong candidate for further preclinical evaluation in treatment of congenital SQT3 and AF.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Marlieke G. Veldhuis
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Jantien Zandvoort
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Fee L. Romunde
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Marien J. C. Houtman
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Karen Duran
- Center for Molecular Medicine, Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gijs van Haaften
- Center for Molecular Medicine, Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Hiroki Takanari
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | | | - Marcel A. G. van der Heyden
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
11
|
Ji Y, Varkevisser R, Opacic D, Bossu A, Kuiper M, Beekman JDM, Yang S, Khan AP, Dobrev D, Voigt N, Wang MZ, Verheule S, Vos MA, van der Heyden MAG. The inward rectifier current inhibitor PA-6 terminates atrial fibrillation and does not cause ventricular arrhythmias in goat and dog models. Br J Pharmacol 2017; 174:2576-2590. [PMID: 28542844 PMCID: PMC5513871 DOI: 10.1111/bph.13869] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 01/24/2023] Open
Abstract
Background and Purpose The density of the inward rectifier current (IK1) increases in atrial fibrillation (AF), shortening effective refractory period and thus promoting atrial re‐entry. The synthetic compound pentamidine analogue 6 (PA‐6) is a selective and potent IK1 inhibitor. We tested PA‐6 for anti‐AF efficacy and potential proarrhythmia, using established models in large animals. Experimental Approach PA‐6 was applied i.v. in anaesthetized goats with rapid pacing‐induced AF and anaesthetized dogs with chronic atrio‐ventricular (AV) block. Electrophysiological and pharmacological parameters were determined. Key Results PA‐6 (2.5 mg·kg−1·10 min−1) induced cardioversion to sinus rhythm (SR) in 5/6 goats and prolonged AF cycle length. AF complexity decreased significantly before cardioversion. PA‐6 accumulated in cardiac tissue with ratios between skeletal muscle : atrial muscle : ventricular muscle of approximately 1:8:21. In SR dogs, PA‐6 peak plasma levels 10 min post infusion were 5.5 ± 0.9 μM, PA‐6 did not induce significant prolongation of QTc and did not affect heart rate, PQ or QRS duration. In dogs with chronic AV block, PA‐6 did not affect QRS but lengthened QTc during the experiment, but not chronically. PA‐6 did not induce TdP arrhythmias in nine animals (0/9) in contrast to dofetilide (5/9). PA‐6 (200 nM) inhibited IK1, but not IK,ACh, in human isolated atrial cardiomyocytes. Conclusion and Implications PA‐6 restored SR in goats with persistent AF and, in dogs with chronic AV block, prolonged QT intervals, without inducing TdP arrhythmias. Our results demonstrate cardiac safety and good anti‐AF properties for PA‐6.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rosanne Varkevisser
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dragan Opacic
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Alexandre Bossu
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marion Kuiper
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Jet D M Beekman
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sihyung Yang
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA
| | - Azinwi Phina Khan
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Niels Voigt
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Michael Zhuo Wang
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | - Marc A Vos
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
12
|
Scherer D, Schworm B, Seyler C, Xynogalos P, Scholz EP, Thomas D, Katus HA, Zitron E. Inhibition of inwardly rectifying Kir2.x channels by the novel anti-cancer agent gambogic acid depends on both pore block and PIP 2 interference. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:701-710. [PMID: 28365825 DOI: 10.1007/s00210-017-1372-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/23/2017] [Indexed: 12/21/2022]
Abstract
The caged xanthone gambogic acid (GA) is a novel anti-cancer agent which exhibits anti-proliferative, anti-inflammatory and cytotoxic effects in many types of cancer tissues. In a recent phase IIa study, GA exhibits a favourable safety profile. However, limited data are available concerning its interaction with cardiac ion channels. Heteromeric assembly of Kir2.x channels underlies the cardiac inwardly rectifying IK1 current which is responsible for the stabilization of the diastolic resting membrane potential. Inhibition of the cardiac IK1 current may lead to ventricular arrhythmia due to delayed afterdepolarizations. Compared to Kv2.1, hERG and Kir1.1, a slow, delayed inhibition of Kir2.1 channels by GA in a mammalian cell line was reported before but no data exist in literature concerning action of GA on homomeric Kir2.2 and Kir2.3 and heteromeric Kir2.x channels. Therefore, the aim of this study was to provide comparative data on the effect of GA on homomeric and heteromeric Kir2.x channels. Homomeric and heteromeric Kir2.x channels were heterologously expressed in Xenopus oocytes, and the two-microelectrode voltage-clamp technique was used to record Kir2.x currents. To investigate the mechanism of the channel inhibition by GA, alanine-mutated Kir2.x channels with modifications in the channels pore region or at phosphatidylinositol 4,5-bisphosphate (PIP2)-binding sites were employed. GA caused a slow inhibition of homomeric and heteromeric Kir2.x channels at low micromolar concentrations (with IC50 Kir2.1/2.2 < Kir2.2 < Kir2.2/2.3 < Kir2.3 < Kir2.1 < Kir2.1/2.3). The effect did not reach saturation within 60 min and was not reversible upon washout for 30 min. The inhibition showed no strong voltage dependence. We provide evidence for a combination of direct channel pore blockade and a PIP2-dependent mechanism as a molecular basis for the observed effect. We conclude that Kir2.x channel inhibition by GA may be relevant in patients with pre-existing cardiac disorders such as chronic heart failure or certain rhythm disorders and recommend a close cardiac monitoring for those patients when treated with GA.
Collapse
Affiliation(s)
- Daniel Scherer
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany.
| | - Benedikt Schworm
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany
| | - Claudia Seyler
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Panagiotis Xynogalos
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany
| | - Eberhard P Scholz
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Edgar Zitron
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| |
Collapse
|
13
|
Inhibition of Kir4.1 potassium channels by quinacrine. Brain Res 2017; 1663:87-94. [PMID: 28288868 DOI: 10.1016/j.brainres.2017.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/24/2017] [Accepted: 03/07/2017] [Indexed: 12/27/2022]
Abstract
Inwardly rectifying potassium (Kir) channels are expressed in many cell types and contribute to a wide range of physiological processes. Particularly, Kir4.1 channels are involved in the astroglial spatial potassium buffering. In this work, we examined the effects of the cationic amphiphilic drug quinacrine on Kir4.1 channels heterologously expressed in HEK293 cells, employing the patch clamp technique. Quinacrine inhibited the currents of Kir4.1 channels in a concentration and voltage dependent manner. In inside-out patches, quinacrine inhibited Kir4.1 channels with an IC50 value of 1.8±0.3μM and with extremely slow blocking and unblocking kinetics. Molecular modeling combined with mutagenesis studies suggested that quinacrine blocks Kir4.1 by plugging the central cavity of the channels, stabilized by the residues E158 and T128. Overall, this study shows that quinacrine blocks Kir4.1 channels, which would be expected to impact the potassium transport in several tissues.
Collapse
|
14
|
Chloroquine blocks the Kir4.1 channels by an open-pore blocking mechanism. Eur J Pharmacol 2017; 800:40-47. [PMID: 28216048 DOI: 10.1016/j.ejphar.2017.02.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/03/2017] [Accepted: 02/15/2017] [Indexed: 11/23/2022]
Abstract
Kir4.1 channels have been implicated in various physiological processes, mainly in the K+ homeostasis of the central nervous system and in the control of glial function and neuronal excitability. Even though, pharmacological research of these channels is very limited. Chloroquine (CQ) is an amino quinolone derivative known to inhibit Kir2.1 and Kir6.2 channels with different action mechanism and binding site. Here, we employed patch-clamp methods, mutagenesis analysis, and molecular modeling to characterize the molecular pharmacology of Kir4.1 inhibition by CQ. We found that this drug inhibits Kir4.1 channels heterologously expressed in HEK-293 cells. CQ produced a fast-onset voltage-dependent pore-blocking effect on these channels. In inside-out patches, CQ showed notable higher potency (IC50 ≈0.5μM at +50mV) and faster onset of block when compared to whole-cell configuration (IC50 ≈7μM at +60mV). Also, CQ showed a voltage-dependent unblock with repolarization. These results suggest that the drug directly blocks Kir4.1 channels by a pore-plugging mechanism. Moreover, we found that two residues (Thr128 and Glu158), facing the central cavity and located within the transmembrane pore, are particularly important structural determinants of CQ block. This evidence was similar to what was previously reported with Kir6.2, but distinct from the interaction site (cytoplasmic pore) CQ-Kir2.1. Thus, our findings highlight the diversity of interaction sites and mechanisms that underlie amino quinolone inhibition of Kir channels.
Collapse
|
15
|
Skarsfeldt MA, Carstensen H, Skibsbye L, Tang C, Buhl R, Bentzen BH, Jespersen T. Pharmacological inhibition of IK1 by PA-6 in isolated rat hearts affects ventricular repolarization and refractoriness. Physiol Rep 2016; 4:4/8/e12734. [PMID: 27117805 PMCID: PMC4848716 DOI: 10.14814/phy2.12734] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/17/2016] [Indexed: 11/24/2022] Open
Abstract
The inwardly rectifying potassium current (IK1) conducted through Kir2.X channels contribute to repolarization of the cardiac action potential and to stabilization of the resting membrane potential in cardiomyocytes. Our aim was to investigate the effect of the recently discovered IK1 inhibitor PA‐6 on action potential repolarization and refractoriness in isolated rat hearts. Transiently transfected HEK‐293 cells expressing IK1 were voltage‐clamped with ramp protocols. Langendorff‐perfused heart experiments were performed on male Sprague–Dawley rats, effective refractory period, Wenckebach cycle length, and ventricular effective refractory period were determined following 200 nmol/L PA‐6 perfusion. 200 nmol/L PA‐6 resulted in a significant time‐latency in drug effect on the IK1 current expressed in HEK‐293 cells, giving rise to a maximal effect at 20 min. In the Langendorff‐perfused heart experiments, PA‐6 prolonged the ventricular action potential duration at 90% repolarization (from 41.8 ± 6.5 msec to 72.6 ± 21.1 msec, 74% compared to baseline, P < 0.01, n = 6). In parallel, PA‐6 significantly prolonged the ventricular effective refractory period compared to baseline (from 34.8 ± 4.6 msec to 58.1 ± 14.7 msec, 67%, P < 0.01, n = 6). PA‐6 increased the short‐term beat‐to‐beat variability and ventricular fibrillation was observed in two of six hearts. Neither atrial ERP nor duration of atrial fibrillation was altered following PA‐6 application. The results show that pharmacological inhibition of cardiac IK1 affects ventricular action potential repolarization and refractoriness and increases the risk of ventricular arrhythmia in isolated rat hearts.
Collapse
Affiliation(s)
- Mark A Skarsfeldt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helena Carstensen
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Skibsbye
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chuyi Tang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Buhl
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bo H Bentzen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Swale DR, Kurata H, Kharade SV, Sheehan J, Raphemot R, Voigtritter KR, Figueroa EE, Meiler J, Blobaum AL, Lindsley CW, Hopkins CR, Denton JS. ML418: The First Selective, Sub-Micromolar Pore Blocker of Kir7.1 Potassium Channels. ACS Chem Neurosci 2016; 7:1013-23. [PMID: 27184474 DOI: 10.1021/acschemneuro.6b00111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The inward rectifier potassium (Kir) channel Kir7.1 (KCNJ13) has recently emerged as a key regulator of melanocortin signaling in the brain, electrolyte homeostasis in the eye, and uterine muscle contractility during pregnancy. The pharmacological tools available for exploring the physiology and therapeutic potential of Kir7.1 have been limited to relatively weak and nonselective small-molecule inhibitors. Here, we report the discovery in a fluorescence-based high-throughput screen of a novel Kir7.1 channel inhibitor, VU714. Site-directed mutagenesis of pore-lining amino acid residues identified glutamate 149 and alanine 150 as essential determinants of VU714 activity. Lead optimization with medicinal chemistry generated ML418, which exhibits sub-micromolar activity (IC50 = 310 nM) and superior selectivity over other Kir channels (at least 17-fold selective over Kir1.1, Kir2.1, Kir2.2, Kir2.3, Kir3.1/3.2, and Kir4.1) except for Kir6.2/SUR1 (equally potent). Evaluation in the EuroFins Lead Profiling panel of 64 GPCRs, ion-channels, and transporters for off-target activity of ML418 revealed a relatively clean ancillary pharmacology. While ML418 exhibited low CLHEP in human microsomes which could be modulated with lipophilicity adjustments, it showed high CLHEP in rat microsomes regardless of lipophilicity. A subsequent in vivo PK study of ML418 by intraperitoneal (IP) administration (30 mg/kg dosage) revealed a suitable PK profile (Cmax = 0.20 μM and Tmax = 3 h) and favorable CNS distribution (mouse brain/plasma Kp of 10.9 to support in vivo studies. ML418, which represents the current state-of-the-art in Kir7.1 inhibitors, should be useful for exploring the physiology of Kir7.1 in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | - Jonathan Sheehan
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | | | | | | | - Jens Meiler
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | | | | | | | | |
Collapse
|
17
|
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
|
18
|
Rodríguez-Menchaca AA, Aréchiga-Figueroa IA, Sánchez-Chapula JA. The molecular basis of chloroethylclonidine block of inward rectifier (Kir2.1 and Kir4.1) K + channels. Pharmacol Rep 2016; 68:383-9. [DOI: 10.1016/j.pharep.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/28/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
|
19
|
van der Heyden MA, Jespersen T. Pharmacological exploration of the resting membrane potential reserve: Impact on atrial fibrillation. Eur J Pharmacol 2016; 771:56-64. [DOI: 10.1016/j.ejphar.2015.11.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/06/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022]
|
20
|
Berger ML, Maciejewska D, Vanden Eynde JJ, Mottamal M, Żabiński J, Kaźmierczak P, Rezler M, Jarak I, Piantanida I, Karminski-Zamola G, Mayence A, Rebernik P, Kumar A, Ismail MA, Boykin DW, Huang TL. Pentamidine analogs as inhibitors of [(3)H]MK-801 and [(3)H]ifenprodil binding to rat brain NMDA receptors. Bioorg Med Chem 2015; 23:4489-4500. [PMID: 26117647 DOI: 10.1016/j.bmc.2015.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 06/05/2015] [Accepted: 06/05/2015] [Indexed: 12/29/2022]
Abstract
The anti-protozoal drug pentamidine is active against opportunistic Pneumocystis pneumonia, but in addition has several other biological targets, including the NMDA receptor (NR). Here we describe the inhibitory potencies of 76 pentamidine analogs at 2 binding sites of the NR, the channel binding site labeled with [(3)H]MK-801 and the [(3)H]ifenprodil binding site. Most analogs acted weaker at the ifenprodil than at the channel site. The spermine-sensitivity of NR inhibition by the majority of the compounds was reminiscent of other long-chain dicationic NR blockers. The potency of the parent compound as NR blocker was increased by modifying the heteroatoms in the bridge connecting the 2 benzamidine moieties and also by integrating the bridge into a seven-membered ring. Docking of the 45 most spermine-sensitive bisbenzamidines to a recently described acidic interface between the N-terminal domains of GluN1 and GluN2B mediating polyamine stimulation of the NR revealed the domain contributed by GluN1 as the most relevant target.
Collapse
Affiliation(s)
- Michael L Berger
- Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | - Dorota Maciejewska
- Department of Organic Chemistry, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Jerzy Żabiński
- Department of Organic Chemistry, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Kaźmierczak
- Department of Organic Chemistry, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Rezler
- Department of Organic Chemistry, Medical University of Warsaw, Warsaw, Poland
| | - Ivana Jarak
- Department of Organic Chemistry, University of Zagreb, Zagreb, Croatia
| | - Ivo Piantanida
- Department of Organic Chemistry, University of Zagreb, Zagreb, Croatia
| | | | - Annie Mayence
- College of Pharmacy, Xavier University of Louisiana, New Orleans, USA
| | - Patrick Rebernik
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Arvind Kumar
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - Mohamed A Ismail
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - David W Boykin
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - Tien L Huang
- College of Pharmacy, Xavier University of Louisiana, New Orleans, USA
| |
Collapse
|
21
|
Gómez R, Caballero R, Barana A, Amorós I, De Palm SH, Matamoros M, Núñez M, Pérez-Hernández M, Iriepa I, Tamargo J, Delpón E. Structural basis of drugs that increase cardiac inward rectifier Kir2.1 currents. Cardiovasc Res 2014; 104:337-46. [PMID: 25205296 DOI: 10.1093/cvr/cvu203] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS We hypothesize that some drugs, besides flecainide, increase the inward rectifier current (IK1) generated by Kir2.1 homotetramers (IKir2.1) and thus, exhibit pro- and/or antiarrhythmic effects particularly at the ventricular level. To test this hypothesis, we analysed the effects of propafenone, atenolol, dronedarone, and timolol on Kir2.x channels. METHODS AND RESULTS Currents were recorded with the patch-clamp technique using whole-cell, inside-out, and cell-attached configurations. Propafenone (0.1 nM-1 µM) did not modify either IK1 recorded in human right atrial myocytes or the current generated by homo- or heterotetramers of Kir2.2 and 2.3 channels recorded in transiently transfected Chinese hamster ovary cells. On the other hand, propafenone increased IKir2.1 (EC50 = 12.0 ± 3.0 nM) as a consequence of its interaction with Cys311, an effect which decreased inward rectification of the current. Propafenone significantly increased mean open time and opening frequency at all the voltages tested, resulting in a significant increase of the mean open probability of the channel. Timolol, which interacted with Cys311, was also able to increase IKir2.1. On the contrary, neither atenolol nor dronedarone modified IKir2.1. Molecular modelling of the Kir2.1-drugs interaction allowed identification of the pharmacophore of drugs that increase IKir2.1. CONCLUSIONS Kir2.1 channels exhibit a binding site determined by Cys311 that is responsible for drug-induced IKir2.1 increase. Drug binding decreases channel affinity for polyamines and current rectification, and can be a mechanism of drug-induced pro- and antiarrhythmic effects not considered until now.
Collapse
Affiliation(s)
- Ricardo Gómez
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Adriana Barana
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Irene Amorós
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Sue-Haida De Palm
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Marcos Matamoros
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Mercedes Núñez
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Marta Pérez-Hernández
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Isabel Iriepa
- Department of Organic Chemistry, School of Pharmacy, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| |
Collapse
|
22
|
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
|
23
|
Xynogalos P, Seyler C, Scherer D, Koepple C, Scholz EP, Thomas D, Katus HA, Zitron E. Class III antiarrhythmic drug dronedarone inhibits cardiac inwardly rectifying Kir2.1 channels through binding at residue E224. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:1153-61. [PMID: 25182566 DOI: 10.1007/s00210-014-1045-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
Abstract
Dronedarone is a novel class III antiarrhythmic drug that is widely used in atrial fibrillation. It has been shown in native cardiomyocytes that dronedarone inhibits cardiac inwardly rectifying current IK1 at high concentrations, which may contribute both its antifibrillatory efficacy and its potential proarrhythmic side effects. However, the underlying mechanism has not been studied in further detail to date. In the mammalian heart, heterotetrameric assembly of Kir2.x channels is the molecular basis of IK1 current. Therefore, we studied the effects of dronedarone on wild-type and mutant Kir2.x channels in the Xenopus oocyte expression system. Dronedarone inhibited Kir2.1 currents but had no effect on Kir2.2 or Kir2.3 currents. Onset of block was slow but completely reversible upon washout. Blockade of Kir2.1 channels did not exhibit strong voltage dependence or frequency dependence. In a screening with different Kir2.1 mutants lacking specific binding sites within the cytoplasmic pore region, we found that residue E224 is essential for binding of dronedarone to Kir2.1 channels. In conclusion, direct block of Kir2.1 channel subunits by dronedarone through binding at E224 may underlie its inhibitory effects on cardiac IK1 current.
Collapse
Affiliation(s)
- Panagiotis Xynogalos
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany,
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Huang TL, Mayence A, Vanden Eynde JJ. Some non-conventional biomolecular targets for diamidines. A short survey. Bioorg Med Chem 2014; 22:1983-92. [DOI: 10.1016/j.bmc.2014.02.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/24/2022]
|
25
|
Swale DR, Kharade SV, Denton JS. Cardiac and renal inward rectifier potassium channel pharmacology: emerging tools for integrative physiology and therapeutics. Curr Opin Pharmacol 2013; 15:7-15. [PMID: 24721648 DOI: 10.1016/j.coph.2013.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/05/2013] [Indexed: 01/01/2023]
Abstract
Inward rectifier potassium (Kir) channels play fundamental roles in cardiac and renal function and may represent unexploited drug targets for cardiovascular diseases. However, the limited pharmacology of Kir channels has slowed progress toward exploring their integrative physiology and therapeutic potential. Here, we review recent progress toward developing the small-molecule pharmacology for Kir2.x, Kir4.1, and Kir7.1 and discuss common mechanistic themes that may help guide future Kir channel-directed drug discovery efforts.
Collapse
Affiliation(s)
- Daniel R Swale
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Sujay V Kharade
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232, United States.
| |
Collapse
|
26
|
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
|
27
|
KCNJ2 mutation in short QT syndrome 3 results in atrial fibrillation and ventricular proarrhythmia. Proc Natl Acad Sci U S A 2013; 110:4291-6. [PMID: 23440193 DOI: 10.1073/pnas.1218154110] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We describe a mutation (E299V) in KCNJ2, the gene that encodes the strong inward rectifier K(+) channel protein (Kir2.1), in an 11-y-old boy. The unique short QT syndrome type-3 phenotype is associated with an extremely abbreviated QT interval (200 ms) on ECG and paroxysmal atrial fibrillation. Genetic screening identified an A896T substitution in a highly conserved region of KCNJ2 that resulted in a de novo mutation E299V. Whole-cell patch-clamp experiments showed that E299V presents an abnormally large outward IK1 at potentials above -55 mV (P < 0.001 versus wild type) due to a lack of inward rectification. Coexpression of wild-type and mutant channels to mimic the heterozygous condition still resulted in a large outward current. Coimmunoprecipitation and kinetic analysis showed that E299V and wild-type isoforms may heteromerize and that their interaction impairs function. The homomeric assembly of E299V mutant proteins actually results in gain of function. Computer simulations of ventricular excitation and propagation using both the homozygous and heterozygous conditions at three different levels of integration (single cell, 2D, and 3D) accurately reproduced the electrocardiographic phenotype of the proband, including an exceedingly short QT interval with merging of the QRS and the T wave, absence of ST segment, and peaked T waves. Numerical experiments predict that, in addition to the short QT interval, absence of inward rectification in the E299V mutation should result in atrial fibrillation. In addition, as predicted by simulations using a geometrically accurate three-dimensional ventricular model that included the His-Purkinje network, a slight reduction in ventricular excitability via 20% reduction of the sodium current should increase vulnerability to life-threatening ventricular tachyarrhythmia.
Collapse
|
28
|
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]
|
29
|
Sirenko O, Crittenden C, Callamaras N, Hesley J, Chen YW, Funes C, Rusyn I, Anson B, Cromwell EF. Multiparameter in vitro assessment of compound effects on cardiomyocyte physiology using iPSC cells. ACTA ACUST UNITED AC 2012; 18:39-53. [PMID: 22972846 DOI: 10.1177/1087057112457590] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A large percentage of drugs fail in clinical studies due to cardiac toxicity; thus, development of sensitive in vitro assays that can evaluate potential adverse effects on cardiomyocytes is extremely important for drug development. Human cardiomyocytes derived from stem cell sources offer more clinically relevant cell-based models than those presently available. Human-induced pluripotent stem cell-derived cardiomyocytes are especially attractive because they express ion channels and demonstrate spontaneous mechanical and electrical activity similar to adult cardiomyocytes. Here we demonstrate techniques for measuring the impact of pharmacologic compounds on the beating rate of cardiomyocytes with ImageXpress Micro and FLIPR Tetra systems. The assays employ calcium-sensitive dyes to monitor changes in Ca(2+) fluxes synchronous with cell beating, which allows monitoring of the beat rate, amplitude, and other parameters. We demonstrate here that the system is able to detect concentration-dependent atypical patterns caused by hERG inhibitors and other ion channel blockers. We also show that both positive and negative chronotropic effects on cardiac rate can be observed and IC(50) values determined. This methodology is well suited for safety testing and can be used to estimate efficacy and dosing of drug candidates prior to clinical studies.
Collapse
|
30
|
Kulzer M, Seyler C, Welke F, Scherer D, Xynogalos P, Scholz EP, Thomas D, Becker R, Karle CA, Katus HA, Zitron E. Inhibition of cardiac Kir2.1–2.3 channels by beta3 adrenoreceptor antagonist SR 59230A. Biochem Biophys Res Commun 2012; 424:315-20. [DOI: 10.1016/j.bbrc.2012.06.114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 06/21/2012] [Indexed: 11/30/2022]
|
31
|
Prole DL, Marrion NV. Identification of putative potassium channel homologues in pathogenic protozoa. PLoS One 2012; 7:e32264. [PMID: 22363819 PMCID: PMC3283738 DOI: 10.1371/journal.pone.0032264] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Accepted: 01/24/2012] [Indexed: 12/21/2022] Open
Abstract
K+ channels play a vital homeostatic role in cells and abnormal activity of these channels can dramatically alter cell function and survival, suggesting that they might be attractive drug targets in pathogenic organisms. Pathogenic protozoa lead to diseases such as malaria, leishmaniasis, trypanosomiasis and dysentery that are responsible for millions of deaths each year worldwide. The genomes of many protozoan parasites have recently been sequenced, allowing rational design of targeted therapies. We analyzed the genomes of pathogenic protozoa and show the existence within them of genes encoding putative homologues of K+ channels. These protozoan K+ channel homologues represent novel targets for anti-parasitic drugs. Differences in the sequences and diversity of human and parasite proteins may allow pathogen-specific targeting of these K+ channel homologues.
Collapse
Affiliation(s)
- David L Prole
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom.
| | | |
Collapse
|
32
|
Dennis AT, Wang L, Wan H, Nassal D, Deschenes I, Ficker E. Molecular determinants of pentamidine-induced hERG trafficking inhibition. Mol Pharmacol 2012; 81:198-209. [PMID: 22046004 PMCID: PMC3263949 DOI: 10.1124/mol.111.075135] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/01/2011] [Indexed: 01/09/2023] Open
Abstract
Pentamidine is an antiprotozoal compound that clinically causes acquired long QT syndrome (acLQTS), which is associated with prolonged QT intervals, tachycardias, and sudden cardiac arrest. Pentamidine delays terminal repolarization in human heart by acutely blocking cardiac inward rectifier currents. At the same time, pentamidine reduces surface expression of the cardiac potassium channel I(Kr)/human ether à-go-go-related gene (hERG). This is unusual in that acLQTS is caused most often by direct block of the cardiac potassium current I(Kr)/hERG. The present study was designed to provide a more complete picture of how hERG surface expression is disrupted by pentamidine at the cellular and molecular levels. Using biochemical and electrophysiological methods, we found that pentamidine exclusively inhibits hERG export from the endoplasmic reticulum to the cell surface in a heterologous expression system as well as in cardiomyocytes. hERG trafficking inhibition could be rescued in the presence of the pharmacological chaperone astemizole. We used rescue experiments in combination with an extensive mutational analysis to locate an interaction site for pentamidine at phenylalanine 656, a crucial residue in the canonical drug binding site of terminally folded hERG. Our data suggest that pentamidine binding to a folding intermediate of hERG arrests channel maturation in a conformational state that cannot be exported from the endoplasmic reticulum. We propose that pentamidine is the founding member of a novel pharmacological entity whose members act as small molecule antichaperones.
Collapse
Affiliation(s)
- Adrienne T Dennis
- Rammelkamp Center for Education and Research, MetroHealth Campus, Cleveland, OH 44109, USA
| | | | | | | | | | | |
Collapse
|
33
|
Houtman MJC, Takanari H, Kok BGJM, van Eck M, Montagne DR, Vos MA, de Boer TP, van der Heyden MAG. Experimental Mapping of the Canine KCNJ2 and KCNJ12 Gene Structures and Functional Analysis of the Canine K(IR)2.2 ion Channel. Front Physiol 2012; 3:9. [PMID: 22363290 PMCID: PMC3277267 DOI: 10.3389/fphys.2012.00009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 01/12/2012] [Indexed: 12/18/2022] Open
Abstract
For many model organisms traditionally in use for cardiac electrophysiological studies, characterization of ion channel genes is lacking. We focused here on two genes encoding the inward rectifier current, KCNJ2 and KCNJ12, in the dog heart. A combination of RT-PCR, 5′-RACE, and 3′-RACE demonstrated the status of KCNJ2 as a two exon gene. The complete open reading frame (ORF) was located on the second exon. One transcription initiation site was mapped. Four differential transcription termination sites were found downstream of two consensus polyadenylation signals. The canine KCNJ12 gene was found to consist of three exons, with its ORF located on the third exon. One transcription initiation and one termination site were found. No alternative splicing was observed in right ventricle or brain cortex. The gene structure of canine KCNJ2 and KCNJ12 was conserved amongst other vertebrates, while current GenBank gene annotation was determined as incomplete. In silico translation of KCN12 revealed a non-conserved glycine rich stretch located near the carboxy-terminus of the KIR2.2 protein. However, no differences were observed when comparing dog with human KIR2.2 protein upon ectopic expression in COS-7 or HEK293 cells with respect to subcellular localization or electrophysiological properties.
Collapse
Affiliation(s)
- Marien J C Houtman
- Division Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht Utrecht, Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Raphemot R, Lonergan DF, Nguyen TT, Utley T, Lewis LM, Kadakia R, Weaver CD, Gogliotti R, Hopkins C, Lindsley CW, Denton JS. Discovery, characterization, and structure-activity relationships of an inhibitor of inward rectifier potassium (Kir) channels with preference for Kir2.3, Kir3.x, and Kir7.1. Front Pharmacol 2011; 2:75. [PMID: 22275899 PMCID: PMC3254186 DOI: 10.3389/fphar.2011.00075] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 11/07/2011] [Indexed: 12/03/2022] Open
Abstract
The inward rectifier family of potassium (Kir) channels is comprised of at least 16 family members exhibiting broad and often overlapping cellular, tissue, or organ distributions. The discovery of disease-causing mutations in humans and experiments on knockout mice has underscored the importance of Kir channels in physiology and in some cases raised questions about their potential as drug targets. However, the paucity of potent and selective small-molecule modulators targeting specific family members has with few exceptions mired efforts to understand their physiology and assess their therapeutic potential. A growing body of evidence suggests that G protein-coupled inward rectifier K (GIRK) channels of the Kir3.X subfamily may represent novel targets for the treatment of atrial fibrillation. In an effort to expand the molecular pharmacology of GIRK, we performed a thallium (Tl(+)) flux-based high-throughput screen of a Kir1.1 inhibitor library for modulators of GIRK. One compound, termed VU573, exhibited 10-fold selectivity for GIRK over Kir1.1 (IC(50) = 1.9 and 19 μM, respectively) and was therefore selected for further study. In electrophysiological experiments performed on Xenopus laevis oocytes and mammalian cells, VU573 inhibited Kir3.1/3.2 (neuronal GIRK) and Kir3.1/3.4 (cardiac GIRK) channels with equal potency and preferentially inhibited GIRK, Kir2.3, and Kir7.1 over Kir1.1 and Kir2.1.Tl(+) flux assays were established for Kir2.3 and the M125R pore mutant of Kir7.1 to support medicinal chemistry efforts to develop more potent and selective analogs for these channels. The structure-activity relationships of VU573 revealed few analogs with improved potency, however two compounds retained most of their activity toward GIRK and Kir2.3 and lost activity toward Kir7.1. We anticipate that the VU573 series will be useful for exploring the physiology and structure-function relationships of these Kir channels.
Collapse
Affiliation(s)
- Rene Raphemot
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
| | - Daniel F. Lonergan
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
| | - Thuy T. Nguyen
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
| | - Thomas Utley
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
| | - L. Michelle Lewis
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
| | - Rishin Kadakia
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
| | - Rocco Gogliotti
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of MedicineNashville, TN, USA
| | - Corey Hopkins
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Chemistry, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Molecular Libraries Probe Production Centers NetworkNashville, TN, USA
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Chemistry, Vanderbilt UniversityNashville, TN, USA
- Vanderbilt Specialized Chemistry Center for Accelerated Probe Development, Molecular Libraries Probe Production Centers NetworkNashville, TN, USA
| | - Jerod S. Denton
- Department of Anesthesiology, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt UniversityNashville, TN, USA
| |
Collapse
|
35
|
Mechanisms for Kir channel inhibition by quinacrine: acute pore block of Kir2.x channels and interference in PIP2 interaction with Kir2.x and Kir6.2 channels. Pflugers Arch 2011; 462:505-17. [DOI: 10.1007/s00424-011-0995-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 07/08/2011] [Accepted: 07/08/2011] [Indexed: 10/18/2022]
|
36
|
Haupt VJ, Schroeder M. Old friends in new guise: repositioning of known drugs with structural bioinformatics. Brief Bioinform 2011; 12:312-26. [DOI: 10.1093/bib/bbr011] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
37
|
van der Heyden MAG, Sánchez-Chapula JA. Toward specific cardiac I(K1) modulators for in vivo application: old drugs point the way. Heart Rhythm 2011; 8:1076-80. [PMID: 21296684 DOI: 10.1016/j.hrthm.2011.01.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 01/26/2011] [Indexed: 10/18/2022]
Affiliation(s)
- Marcel A G van der Heyden
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | |
Collapse
|
38
|
Inhibition of lysosomal degradation rescues pentamidine-mediated decreases of KIR2.1 ion channel expression but not that of Kv11.1. Eur J Pharmacol 2011; 652:96-103. [DOI: 10.1016/j.ejphar.2010.10.093] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 10/13/2010] [Accepted: 10/29/2010] [Indexed: 11/19/2022]
|
39
|
de Boer TP, Houtman MJC, Compier M, van der Heyden MAG. The mammalian K(IR)2.x inward rectifier ion channel family: expression pattern and pathophysiology. Acta Physiol (Oxf) 2010; 199:243-56. [PMID: 20331539 DOI: 10.1111/j.1748-1716.2010.02108.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Inward rectifier currents based on K(IR)2.x subunits are regarded as essential components for establishing a stable and negative resting membrane potential in many excitable cell types. Pharmacological inhibition, null mutation in mice and dominant positive and negative mutations in patients reveal some of the important functions of these channels in their native tissues. Here we review the complex mammalian expression pattern of K(IR)2.x subunits and relate these to the outcomes of functional inhibition of the resultant channels. Correlations between expression and function in muscle and bone tissue are observed, while we recognize a discrepancy between neuronal expression and function.
Collapse
Affiliation(s)
- T P de Boer
- Department of Medical Physiology, UMCU, Utrecht, the Netherlands
| | | | | | | |
Collapse
|