1
|
Zha T, Fang X, Wan J, Chen X, Lin J, Chen Q. Preclinical Insights into the Role of Kir4.1 in Chronic Pain and Depression: Mechanisms and Therapeutic Potential. Biomolecules 2025; 15:165. [PMID: 40001468 PMCID: PMC11852603 DOI: 10.3390/biom15020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic pain and mental health disorders, such as depression and anxiety, frequently co-occur and share underlying mechanisms involving neuronal excitability and synaptic transmission. The inwardly rectifying potassium channel 4.1 (Kir4.1), predominantly expressed in glial cells, is crucial for maintaining extracellular potassium and glutamate homeostasis. Dysregulation of Kir4.1 leads to altered neuronal activity, contributing to both chronic pain and mental health disorders. In chronic pain, downregulation of Kir4.1 impairs potassium buffering and glutamate clearance, increasing neuronal excitability and enhancing pain signaling through peripheral and central sensitization. In mental health disorders, impaired Kir4.1 function disrupts neurotrophic factor secretion and neuroinflammatory pathways, leading to mood disturbances. This review primarily summarizes findings from preclinical studies to examine the relationship between Kir4.1 and the pathogenesis of chronic pain and mental health disorders, discussing its molecular structure, expression patterns, and functional roles. Furthermore, we explore therapeutic strategies targeting Kir4.1, including pharmacological modulators and gene therapy approaches, emphasizing its potential as a novel therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | - Jiu Lin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310006, China; (T.Z.); (X.F.); (J.W.); (X.C.)
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310006, China; (T.Z.); (X.F.); (J.W.); (X.C.)
| |
Collapse
|
2
|
Pitsillou E, Liang JJ, Kino N, Lockwood JL, Hung A, El-Osta A, AbuMaziad AS, Karagiannis TC. An In Silico Investigation of the Pathogenic G151R G Protein-Gated Inwardly Rectifying K + Channel 4 Variant to Identify Small Molecule Modulators. BIOLOGY 2024; 13:992. [PMID: 39765659 PMCID: PMC11727529 DOI: 10.3390/biology13120992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 01/15/2025]
Abstract
Primary aldosteronism is characterised by the excessive production of aldosterone, which is a key regulator of salt metabolism, and is the most common cause of secondary hypertension. Studies have investigated the association between primary aldosteronism and genetic alterations, with pathogenic mutations being identified. This includes a glycine-to-arginine substitution at position 151 (G151R) of the G protein-activated inward rectifier potassium (K+) channel 4 (GIRK4), which is encoded by the KCNJ5 gene. Mutations in GIRK4 have been found to reduce the selectivity for K+ ions, resulting in membrane depolarisation, the activation of voltage-gated Ca2+ channels, and an increase in aldosterone secretion. As a result, there is an interest in identifying and exploring the mechanisms of action of small molecule modulators of wildtype (WT) and mutant channels. In order to investigate the potential modulation of homotetrameric GIRK4WT and GIRK4G151R channels, homology models were generated. Molecular dynamics (MD) simulations were performed, followed by a cluster analysis to extract starting structures for molecular docking. The central cavity has been previously identified as a binding site for small molecules, including natural compounds. The OliveNetTM database, which consists of over 600 compounds from Olea europaea, was subsequently screened against the central cavity. The binding affinities and interactions of the docked ligands against the GIRK4WT and GIRK4G151R channels were then examined. Based on the results, luteolin-7-O-rutinoside, pheophorbide a, and corosolic acid were identified as potential lead compounds. The modulatory activity of olive-derived compounds against the WT and mutated forms of the GIRK4 channel can be evaluated further in vitro.
Collapse
Affiliation(s)
- Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED Polytechnic, Melbourne, VIC 3053, Australia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Julia J. Liang
- Epigenomic Medicine Laboratory at prospED Polytechnic, Melbourne, VIC 3053, Australia
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Noa Kino
- Epigenomic Medicine Laboratory at prospED Polytechnic, Melbourne, VIC 3053, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jessica L. Lockwood
- Epigenomic Medicine Laboratory at prospED Polytechnic, Melbourne, VIC 3053, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30-32 Ngan Shing Street, Sha Tin, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
- Biomedical Laboratory Science, Department of Technology, Faculty of Health, University College Copenhagen, 2200 Copenhagen, Denmark
| | - Asmaa S. AbuMaziad
- Department of Pediatrics, College of Medicine Tucson, The University of Arizona, Tucson, AZ 85724, USA
| | - Tom C. Karagiannis
- Epigenomic Medicine Laboratory at prospED Polytechnic, Melbourne, VIC 3053, Australia
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
3
|
Janjic P, Solev D, Kocarev L. Non-trivial dynamics in a model of glial membrane voltage driven by open potassium pores. Biophys J 2023; 122:1470-1490. [PMID: 36919241 PMCID: PMC10147837 DOI: 10.1016/j.bpj.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/01/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Despite the molecular evidence that a nearly linear steady-state current-voltage relationship in mammalian astrocytes reflects a total current resulting from more than one differentially regulated K+ conductance, detailed ordinary differential equation (ODE) models of membrane voltage Vm are still lacking. Various experimental results reporting altered rectification of the major Kir currents in glia, dominated by Kir4.1, have motivated us to develop a detailed model of Vm dynamics incorporating the weaker potassium K2P-TREK1 current in addition to Kir4.1, and study the stability of the resting state Vr. The main question is whether, with the loss of monotonicity in glial I-V curve resulting from altered Kir rectification, the nominal resting state Vr remains stable, and the cell retains the trivial, potassium electrode behavior with Vm after EK. The minimal two-dimensional model of Vm near Vr showed that an N-shape deformed Kir I-V curve induces multistability of Vm in a model that incorporates K2P activation kinetics, and nonspecific K+ leak currents. More specifically, an asymmetrical, nonlinear decrease of outward Kir4.1 conductance, turning the channels into inward rectifiers, introduces instability of Vr. That happens through a robust bifurcation giving birth to a second, more depolarized stable resting state Vdr > -10 mV. Realistic recordings from electrographic seizures were used to perturb the model. Simulations of the model perturbed by constant current through gap junctions and seizure-like discharges as local field potentials led to depolarization and switching of Vm between the two stable states, in a downstate-upstate manner. In the event of prolonged depolarizations near Vdr, such catastrophic instability would affect all aspects of the glial function, from metabolic support to membrane transport, and practically all neuromodulatory roles assigned to glia.
Collapse
Affiliation(s)
- Predrag Janjic
- Laboratory for Complex Systems and Networks, Research Centre for Computer Science and Information Technologies, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia.
| | - Dimitar Solev
- Laboratory for Complex Systems and Networks, Research Centre for Computer Science and Information Technologies, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| | - Ljupco Kocarev
- Laboratory for Complex Systems and Networks, Research Centre for Computer Science and Information Technologies, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| |
Collapse
|
4
|
Costa EA, Gonçalves AP, Batista JAD, Bazoni RF, Santos AA, Rocha MS. New Insights into the Mechanism of Action of the Drug Chloroquine: Direct Interaction with DNA and Cytotoxicity. J Phys Chem B 2022; 126:3512-3521. [PMID: 35533378 DOI: 10.1021/acs.jpcb.2c01119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chloroquine (CLQ) and hydroxychloroquine (HCLQ) are compounds largely employed in the treatment of various human diseases for decades. Nevertheless, a number of intrinsic details concerning their mechanisms of action, especially at the molecular level, are still unknown or have presented controversial results in the literature. Using optical tweezers, here, we investigate at the single-molecule level the molecular mechanism of action of the drug CLQ in its intrinsic interaction with the double-stranded (ds)DNA molecule, one of its targets inside cells, determining the binding modes and the physicochemical (binding) parameters of the interaction. In particular, we show that the ionic strength of the surrounding medium strongly influences such interaction, changing even the main binding mode. In addition, the cytotoxicity of CLQ against three different cell lines was also investigated here, allowing one to evaluate and compare the effect of the drug on the cell viability. In particular, we show that CLQ is highly cytotoxic at a very low (a few micromolar) concentration range for all cell lines tested. These results were rigorously compared to the equivalent ones obtained for the closely related compound hydroxychloroquine (HCLQ), allowing a critical comparison between the action of these drugs at the molecular and cellular levels.
Collapse
Affiliation(s)
- Ethe A Costa
- Departamento de Física, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil
| | - Amanda P Gonçalves
- Departamento de Biologia Geral, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil
| | - Josiane A D Batista
- Departamento de Física, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36.036-900, Brazil
| | - Raniella F Bazoni
- Departamento de Ciências Naturais, Universidade Federal do Espírito Santo, São Mateus, Espírito Santo 29.932-900, Brazil
| | - Anésia A Santos
- Departamento de Biologia Geral, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil
| | - Márcio S Rocha
- Departamento de Física, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil
| |
Collapse
|
5
|
McClenahan SJ, Kent CN, Kharade SV, Isaeva E, Williams JC, Han C, Terker A, Gresham R, Lazarenko RM, Days EL, Romaine IM, Bauer JA, Boutaud O, Sulikowski GA, Harris R, Weaver CD, Staruschenko A, Lindsley CW, Denton JS. VU6036720: The First Potent and Selective In Vitro Inhibitor of Heteromeric Kir4.1/5.1 Inward Rectifier Potassium Channels. Mol Pharmacol 2022; 101:357-370. [PMID: 35246480 PMCID: PMC9092466 DOI: 10.1124/molpharm.121.000464] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/14/2022] [Indexed: 01/14/2023] Open
Abstract
Heteromeric Kir4.1/Kir5.1 (KCNJ10/KCNJ16) inward rectifier potassium (Kir) channels play key roles in the brain and kidney, but pharmacological tools for probing their physiology and therapeutic potential have not been developed. Here, we report the discovery, in a high-throughput screening of 80,475 compounds, of the moderately potent and selective inhibitor VU0493690, which we selected for characterization and chemical optimization. VU0493690 concentration-dependently inhibits Kir4.1/5.1 with an IC50 of 0.96 μM and exhibits at least 10-fold selectivity over Kir4.1 and ten other Kir channels. Multidimensional chemical optimization of VU0493690 led to the development of VU6036720, the most potent (IC50 = 0.24 μM) and selective (>40-fold over Kir4.1) Kir4.1/5.1 inhibitor reported to date. Cell-attached patch single-channel recordings revealed that VU6036720 inhibits Kir4.1/5.1 activity through a reduction of channel open-state probability and single-channel current amplitude. Elevating extracellular potassium ion by 20 mM shifted the IC50 6.8-fold, suggesting that VU6036720 is a pore blocker that binds in the ion-conduction pathway. Mutation of the "rectification controller" asparagine 161 to glutamate (N161E), which is equivalent to small-molecule binding sites in other Kir channels, led to a strong reduction of inhibition by VU6036720. Renal clearance studies in mice failed to show a diuretic response that would be consistent with inhibition of Kir4.1/5.1 in the renal tubule. Drug metabolism and pharmacokinetics profiling revealed that high VU6036720 clearance and plasma protein binding may prevent target engagement in vivo. In conclusion, VU6036720 represents the current state-of-the-art Kir4.1/5.1 inhibitor that should be useful for probing the functions of Kir4.1/5.1 in vitro and ex vivo. SIGNIFICANCE STATEMENT: Heteromeric inward rectifier potassium (Kir) channels comprising Kir4.1 and Kir5.1 subunits play important roles in renal and neural physiology and may represent inhibitory drug targets for hypertension and edema. Herein, we employ high-throughput compound library screening, patch clamp electrophysiology, and medicinal chemistry to develop and characterize the first potent and specific in vitro inhibitor of Kir4.1/5.1, VU6036720, which provides proof-of-concept that drug-like inhibitors of this channel may be developed.
Collapse
Affiliation(s)
- Samantha J McClenahan
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Caitlin N Kent
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Sujay V Kharade
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Elena Isaeva
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Jade C Williams
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Changho Han
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Andrew Terker
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Robert Gresham
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Roman M Lazarenko
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Emily L Days
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Ian M Romaine
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Joshua A Bauer
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Olivier Boutaud
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Gary A Sulikowski
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Raymond Harris
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - C David Weaver
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Alexander Staruschenko
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Craig W Lindsley
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| | - Jerod S Denton
- Departments of Anesthesiology (S.J.M., S.V.K., R.G., R.M.L., J.S.D.), Biochemistry (J.A.B.), Chemistry (C.N.K., J.C.W., I.M.R., C.D.W., G.A.S., C.W.L.), Pharmacology (E.L.D., C.D.W., C.W.L., C.H., O.B., J.S.D.), and Nephrology (A.T., R.H.), and Vanderbilt Institute of Chemical Biology (J.A.B., G.S., C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin (E.I.); and Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida (A.S.)
| |
Collapse
|
6
|
Tarantino N, Della Rocca DG, Zou F, Lin A, Natale A, Di Biase L. Prevalence, Outcomes, and Management of Ventricular Arrhythmias in COVID-19 Patients. Card Electrophysiol Clin 2022; 14:11-20. [PMID: 35221078 PMCID: PMC8554003 DOI: 10.1016/j.ccep.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We review the current data on epidemiology, the clinical significance, the pathophysiologic mechanisms, and the treatment of VAs in the setting of COVID-19. VAs prevail in 0.15% to 8% of hospitalized patients, but only sustained and rapid tachyarrhythmias are purportedly associated with a significant increase in mortality. Multiple factors can elicit VAs, which are ultimately deemed to be a marker of severe systemic disease rather than a distinct cardiac condition. Even though the electrophysiologist plays a determinant role in the secondary prevention of VAs, a multidisciplinary approach is indispensable for primary prophylaxis and acute management.
Collapse
Affiliation(s)
- Nicola Tarantino
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA
| | - Domenico G Della Rocca
- Texas Cardiac Arrhythmia Institute at St. David's Medical Center, 3000 N I-35, Suite 720, Austin, TX 78705, USA
| | - Fengwei Zou
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA
| | - Aung Lin
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA
| | - Andrea Natale
- Texas Cardiac Arrhythmia Institute at St. David's Medical Center, 3000 N I-35, Suite 720, Austin, TX 78705, USA; Scripps Interventional Car, 9834 Genesee Ave, La Jolla, CA 92037, USA; Health Education Campus, 9501 Euclid Ave, Cleveland, OH 44106, USA
| | - Luigi Di Biase
- Montefiore Medical Center, 111 E 210th street, Bronx, NY 10467, USA; Texas Cardiac Arrhythmia Institute at St. David's Medical Center, 3000 N I-35, Suite 720, Austin, TX 78705, USA.
| |
Collapse
|
7
|
Ohno Y, Kunisawa N, Shimizu S. Emerging Roles of Astrocyte Kir4.1 Channels in the Pathogenesis and Treatment of Brain Diseases. Int J Mol Sci 2021; 22:ijms221910236. [PMID: 34638578 PMCID: PMC8508600 DOI: 10.3390/ijms221910236] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/15/2022] Open
Abstract
Inwardly rectifying Kir4.1 channels in astrocytes mediate spatial potassium (K+) buffering, a clearance mechanism for excessive extracellular K+, in tripartite synapses. In addition to K+ homeostasis, astrocytic Kir4.1 channels also play an essential role in regulating extracellular glutamate levels via coupling with glutamate transporters. Moreover, Kir4.1 channels act as novel modulators of the expression of brain-derived neurotrophic factor (BDNF) in astrocytes. Specifically, inhibition of astrocytic Kir4.1 channels elevates extracellular K+ and glutamate levels at synapses and facilitates BDNF expression in astrocytes. These changes elevate neural excitability, which may facilitate synaptic plasticity and connectivity. In this article, we summarize the functions and pharmacological features of Kir4.1 channels in astrocytes and highlight the importance of these channels in the treatment of brain diseases. Although further validation in animal models and human patients is required, astrocytic Kir4.1 channel could potentially serve as a novel therapeutic target for the treatment of depressive disorders and epilepsy.
Collapse
|
8
|
Kir Channel Molecular Physiology, Pharmacology, and Therapeutic Implications. Handb Exp Pharmacol 2021; 267:277-356. [PMID: 34345939 DOI: 10.1007/164_2021_501] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
For the past two decades several scholarly reviews have appeared on the inwardly rectifying potassium (Kir) channels. We would like to highlight two efforts in particular, which have provided comprehensive reviews of the literature up to 2010 (Hibino et al., Physiol Rev 90(1):291-366, 2010; Stanfield et al., Rev Physiol Biochem Pharmacol 145:47-179, 2002). In the past decade, great insights into the 3-D atomic resolution structures of Kir channels have begun to provide the molecular basis for their functional properties. More recently, computational studies are beginning to close the time domain gap between in silico dynamic and patch-clamp functional studies. The pharmacology of these channels has also been expanding and the dynamic structural studies provide hope that we are heading toward successful structure-based drug design for this family of K+ channels. In the present review we focus on placing the physiology and pharmacology of this K+ channel family in the context of atomic resolution structures and in providing a glimpse of the promising future of therapeutic opportunities.
Collapse
|
9
|
Kinboshi M, Ikeda A, Ohno Y. Role of Astrocytic Inwardly Rectifying Potassium (Kir) 4.1 Channels in Epileptogenesis. Front Neurol 2020; 11:626658. [PMID: 33424762 PMCID: PMC7786246 DOI: 10.3389/fneur.2020.626658] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/08/2020] [Indexed: 12/25/2022] Open
Abstract
Astrocytes regulate potassium and glutamate homeostasis via inwardly rectifying potassium (Kir) 4.1 channels in synapses, maintaining normal neural excitability. Numerous studies have shown that dysfunction of astrocytic Kir4.1 channels is involved in epileptogenesis in humans and animal models of epilepsy. Specifically, Kir4.1 channel inhibition by KCNJ10 gene mutation or expressional down-regulation increases the extracellular levels of potassium ions and glutamate in synapses and causes hyperexcitation of neurons. Moreover, recent investigations demonstrated that inhibition of Kir4.1 channels facilitates the expression of brain-derived neurotrophic factor (BDNF), an important modulator of epileptogenesis, in astrocytes. In this review, we summarize the current understanding on the role of astrocytic Kir4.1 channels in epileptogenesis, with a focus on functional and expressional changes in Kir4.1 channels and their regulation of BDNF secretion. We also discuss the potential of Kir4.1 channels as a therapeutic target for the prevention of epilepsy.
Collapse
Affiliation(s)
- Masato Kinboshi
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, Japan.,Department of Epilepsy, Movement Disorders and Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akio Ikeda
- Department of Epilepsy, Movement Disorders and Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukihiro Ohno
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, Japan
| |
Collapse
|
10
|
Mubagwa K. Cardiac effects and toxicity of chloroquine: a short update. Int J Antimicrob Agents 2020; 56:106057. [PMID: 32565195 PMCID: PMC7303034 DOI: 10.1016/j.ijantimicag.2020.106057] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/07/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023]
Abstract
There is currently increased interest in the use of the antimalarial drugs chloroquine and hydroxychloroquine for the treatment of other diseases, including cancer and viral infections such as coronavirus disease 2019 (COVID-19). However, the risk of cardiotoxic effects tends to limit their use. In this review, the effects of these drugs on the electrical and mechanical activities of the heart as well as on remodelling of cardiac tissue are presented and the underlying molecular and cellular mechanisms are discussed. The drugs can have proarrhythmic as well as antiarrhythmic actions resulting from their inhibition of ion channels, including voltage-dependent Na+ and Ca2+ channels, background and voltage-dependent K+ channels, and pacemaker channels. The drugs also exert a vagolytic effect due at least in part to a muscarinic receptor antagonist action. They also interfere with normal autophagy flux, an effect that could aggravate ischaemia/reperfusion injury or post-infarct remodelling. Most of the toxic effects occur at high concentrations, following prolonged drug administration or in the context of drug associations.
Collapse
Affiliation(s)
- Kanigula Mubagwa
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Basic Sciences, Faculty of Medicine, Université Catholique de Bukavu, Bukavu, DR Congo.
| |
Collapse
|
11
|
Morán-Zendejas R, Delgado-Ramírez M, Xu J, Valdés-Abadía B, Aréchiga-Figueroa IA, Cui M, Rodríguez-Menchaca AA. In vitro and in silico characterization of the inhibition of Kir4.1 channels by aminoglycoside antibiotics. Br J Pharmacol 2020; 177:4548-4560. [PMID: 32726456 DOI: 10.1111/bph.15214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/11/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Aminoglycoside antibiotics are positively charged molecules that are known to inhibit several ion channels. In this study, we have shown that aminoglycosides also inhibit the activity of Kir4.1 channels. Aminoglycosides inhibit Kir4.1 channels by a pore-blocking mechanism, plugging the central vestibule of the channel. EXPERIMENTAL APPROACH Patch-clamp recordings were made in HEK-293 cells transiently expressing Kir4.1 channels to analyse the effects of gentamicin, neomycin and kanamycin. In silico modelling followed by mutagenesis were realized to identify the residues critical for aminoglycosides binding to Kir4.1. KEY RESULTS Aminoglycoside antibiotics block Kir4.1 channels in a concentration- and voltage-dependent manner, getting access to the protein from the intracellular side of the plasma membrane. Aminoglycosides block Ki4.1 with a rank order of potency as follows: gentamicin ˃ neomycin ˃ kanamycin. The residues T128 and principally E158, facing the central cavity of Kir4.1, are important structural determinants for aminoglycosides binding to the channel, as determined by our in silico modelling and confirmed by mutagenesis experiments. CONCLUSION AND IMPLICATIONS Kir4.1 channels are also target of aminoglycoside antibiotics, which could affect potassium transport in several tissues.
Collapse
Affiliation(s)
- Rita Morán-Zendejas
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Mayra Delgado-Ramírez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Jie Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Belkis Valdés-Abadía
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Aldo A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| |
Collapse
|
12
|
McAlinden KD, Deshpande DA, Ghavami S, Xenaki D, Sohal SS, Oliver BG, Haghi M, Sharma P. Autophagy Activation in Asthma Airways Remodeling. Am J Respir Cell Mol Biol 2019; 60:541-553. [PMID: 30383396 DOI: 10.1165/rcmb.2018-0169oc] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Current asthma therapies fail to target airway remodeling that correlates with asthma severity driving disease progression that ultimately leads to loss of lung function. Macroautophagy (hereinafter "autophagy") is a fundamental cell-recycling mechanism in all eukaryotic cells; emerging evidence suggests that it is dysregulated in asthma. We investigated the interrelationship between autophagy and airway remodeling and assessed preclinical efficacy of a known autophagy inhibitor in murine models of asthma. Human asthmatic and nonasthmatic lung tissues were histologically evaluated and were immunostained for key autophagy markers. The percentage area of positive staining was quantified in the epithelium and airway smooth muscle bundles using ImageJ software. Furthermore, the autophagy inhibitor chloroquine was tested intranasally in prophylactic (3 wk) and treatment (5 wk) models of allergic asthma in mice. Human asthmatic tissues showed greater tissue inflammation and demonstrated hallmark features of airway remodeling, displaying thickened epithelium (P < 0.001) and reticular basement membrane (P < 0.0001), greater lamina propria depth (P < 0.005), and increased airway smooth muscle bundles (P < 0.001) with higher expression of Beclin-1 (P < 0.01) and ATG5 (autophagy-related gene 5) (P < 0.05) together with reduced p62 (P < 0.05) compared with nonasthmatic control tissues. Beclin-1 expression was significantly higher in asthmatic epithelium and ciliated cells (P < 0.05), suggesting a potential role of ciliophagy in asthma. Murine asthma models demonstrated effective preclinical efficacy (reduced key features of allergic asthma: airway inflammation, airway hyperresponsiveness, and airway remodeling) of the autophagy inhibitor chloroquine. Our data demonstrate cell context-dependent and selective activation of autophagy in structural cells in asthma. Furthermore, this pathway can be effectively targeted to ameliorate airway remodeling in asthma.
Collapse
Affiliation(s)
- Kielan D McAlinden
- 1 Graduate School of Health and.,3 School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,2 Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Deepak A Deshpande
- 4 Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Saeid Ghavami
- 5 Department of Anatomy & Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - Dia Xenaki
- 2 Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Sukhwinder Singh Sohal
- 6 Respiratory Translational Research Group, Department of Laboratory Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Brian G Oliver
- 3 School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,2 Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | | | - Pawan Sharma
- 3 School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,2 Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Chloroquine inhibits tumor-related Kv10.1 channel and decreases migration of MDA-MB-231 breast cancer cells in vitro. Eur J Pharmacol 2019; 855:262-266. [PMID: 31082369 DOI: 10.1016/j.ejphar.2019.05.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/05/2019] [Accepted: 05/06/2019] [Indexed: 12/25/2022]
Abstract
Chloroquine (CQ) is an old antimalarial drug currently being investigated for its anti-tumor properties. As chloroquine has been shown to inhibits several potassium channels, we decided to study its effect on the tumor-related Kv10.1 channel by using patch-clamp electrophysiology and cell migration assays. We found that chloroquine inhibited Kv10.1 channels transiently expressed in HEK-293 cells in a concentration- and voltage-dependent manner acting from the cytoplasmic side of the plasma membrane. Chloroquine also inhibited the outward potassium currents from MDA-MB-231 cells, which are mainly carried through Kv10.1 channels as was confirmed using astemizole. Additionally, chloroquine decreased MDA-MB-231 cell migration in the in vitro scratch wound healing assay. In conclusion, our data suggest that chloroquine decreases MDA-MB-231 cell migration by inhibiting Kv10.1 channels. The inhibition of Kv10.1 channels could represent another mechanism of the antitumoral action of chloroquine, besides autophagy inhibition and tumor vessel normalization.
Collapse
|
14
|
Ohno Y, Kinboshi M, Shimizu S. Inwardly Rectifying Potassium Channel Kir4.1 as a Novel Modulator of BDNF Expression in Astrocytes. Int J Mol Sci 2018; 19:ijms19113313. [PMID: 30356026 PMCID: PMC6274740 DOI: 10.3390/ijms19113313] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/02/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a key molecule essential for neural plasticity and development, and is implicated in the pathophysiology of various central nervous system (CNS) disorders. It is now documented that BDNF is synthesized not only in neurons, but also in astrocytes which actively regulate neuronal activities by forming tripartite synapses. Inwardly rectifying potassium (Kir) channel subunit Kir4.1, which is specifically expressed in astrocytes, constructs Kir4.1 and Kir4.1/5.1 channels, and mediates the spatial potassium (K+) buffering action of astrocytes. Recent evidence illustrates that Kir4.1 channels play important roles in bringing about the actions of antidepressant drugs and modulating BDNF expression in astrocytes. Although the precise mechanisms remain to be clarified, it seems likely that inhibition (down-regulation or blockade) of astrocytic Kir4.1 channels attenuates K+ buffering, increases neuronal excitability by elevating extracellular K+ and glutamate, and facilitates BDNF expression. Conversely, activation (up-regulation or opening) of Kir4.1 channels reduces neuronal excitability by lowering extracellular K+ and glutamate, and attenuates BDNF expression. Particularly, the former pathophysiological alterations seem to be important in epileptogenesis and pain sensitization, and the latter in the pathogenesis of depressive disorders. In this article, we review the functions of Kir4.1 channels, with a focus on their regulation of spatial K+ buffering and BDNF expression in astrocytes, and discuss the role of the astrocytic Kir4.1-BDNF system in modulating CNS disorders.
Collapse
Affiliation(s)
- Yukihiro Ohno
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Masato Kinboshi
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | - Saki Shimizu
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| |
Collapse
|
15
|
Kharade SV, Kurata H, Bender AM, Blobaum AL, Figueroa EE, Duran A, Kramer M, Days E, Vinson P, Flores D, Satlin LM, Meiler J, Weaver CD, Lindsley CW, Hopkins CR, Denton JS. Discovery, Characterization, and Effects on Renal Fluid and Electrolyte Excretion of the Kir4.1 Potassium Channel Pore Blocker, VU0134992. Mol Pharmacol 2018; 94:926-937. [PMID: 29895592 PMCID: PMC6041953 DOI: 10.1124/mol.118.112359] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/30/2018] [Indexed: 12/28/2022] Open
Abstract
The inward rectifier potassium (Kir) channel Kir4.1 (KCNJ10) carries out important physiologic roles in epithelial cells of the kidney, astrocytes in the central nervous system, and stria vascularis of the inner ear. Loss-of-function mutations in KCNJ10 lead to EAST/SeSAME syndrome, which is characterized by epilepsy, ataxia, renal salt wasting, and sensorineural deafness. Although genetic approaches have been indispensable for establishing the importance of Kir4.1 in the normal function of these tissues, the availability of pharmacological tools for acutely manipulating the activity of Kir4.1 in genetically normal animals has been lacking. We therefore carried out a high-throughput screen of 76,575 compounds from the Vanderbilt Institute of Chemical Biology library for small-molecule modulators of Kir4.1. The most potent inhibitor identified was 2-(2-bromo-4-isopropylphenoxy)-N-(2,2,6,6-tetramethylpiperidin-4-yl)acetamide (VU0134992). In whole-cell patch-clamp electrophysiology experiments, VU0134992 inhibits Kir4.1 with an IC50 value of 0.97 µM and is 9-fold selective for homomeric Kir4.1 over Kir4.1/5.1 concatemeric channels (IC50 = 9 µM) at -120 mV. In thallium (Tl+) flux assays, VU0134992 is greater than 30-fold selective for Kir4.1 over Kir1.1, Kir2.1, and Kir2.2; is weakly active toward Kir2.3, Kir6.2/SUR1, and Kir7.1; and is equally active toward Kir3.1/3.2, Kir3.1/3.4, and Kir4.2. This potency and selectivity profile is superior to Kir4.1 inhibitors amitriptyline, nortriptyline, and fluoxetine. Medicinal chemistry identified components of VU0134992 that are critical for inhibiting Kir4.1. Patch-clamp electrophysiology, molecular modeling, and site-directed mutagenesis identified pore-lining glutamate 158 and isoleucine 159 as critical residues for block of the channel. VU0134992 displayed a large free unbound fraction (fu) in rat plasma (fu = 0.213). Consistent with the known role of Kir4.1 in renal function, oral dosing of VU0134992 led to a dose-dependent diuresis, natriuresis, and kaliuresis in rats. Thus, VU0134992 represents the first in vivo active tool compound for probing the therapeutic potential of Kir4.1 as a novel diuretic target for the treatment of hypertension.
Collapse
Affiliation(s)
- Sujay V Kharade
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Haruto Kurata
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Aaron M Bender
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Anna L Blobaum
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Eric E Figueroa
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Amanda Duran
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Meghan Kramer
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Emily Days
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Paige Vinson
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Daniel Flores
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Lisa M Satlin
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Jens Meiler
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - C David Weaver
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Craig W Lindsley
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Corey R Hopkins
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee (S.V.K., M.K., J.S.D.); Center for Neuroscience Drug Discovery and the Vanderbilt Specialized Chemistry Center for Accelerated Probe Development (H.K., A.M.B., A.L.B., C.W.L., C.R.H.), Departments of Pharmacology (H.K., A.M.B., E.E.F., J.M., C.D.W., C.W.L., J.S.D.) and Chemistry (A.D., J.M., C.D.W., C.W.L.), High-Throughput Screening Center (E.D., P.V.), and Institute of Chemical Biology (C.D.W., C.W.L., J.S.D.), Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York (D.F., L.M.S.); and Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.)
| |
Collapse
|
16
|
Inanobe A, Itamochi H, Kurachi Y. Kir Channel Blockages by Proflavine Derivatives via Multiple Modes of Interaction. Mol Pharmacol 2018; 93:592-600. [PMID: 29650538 DOI: 10.1124/mol.117.111377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/06/2018] [Indexed: 11/22/2022] Open
Abstract
Many compounds inhibit tetrameric and pseudo-tetrameric cation channels by associating with the central cavity located in the middle of the membrane plane. They traverse the ion conduction pathway from the intracellular side and through access to the cavity. Previously, we reported that the bacteriostatic agent, proflavine, preferentially blocked a subset of inward rectifier K+ (Kir) channels. However, the development of the inhibition of Kir1.1 by the compound was obviously different from that operating in Kir3.2 as a pore blocker. To gain mechanistic insights into the compound-channel interaction, we analyzed its chemical specificity, subunit selectivity, and voltage dependency using 13 different combinations of Kir-channel family members and 11 proflavine derivatives. The Kir-channel family members were classified into three groups: 1) Kir2.2, Kir3.x, Kir4.2, and Kir6.2Δ36, which exhibited Kir3.2-type inhibition (slow onset and recovery, irreversible, and voltage-dependent blockage); 2) Kir1.1 and Kir4.1/Kir5.1 (prompt onset and recovery, reversible, and voltage-independent blockage); and 3) Kir2.1, Kir2.3, Kir4.1, and Kir7.1 (no response). The degree of current inhibition depended on the combination of compounds and channels. Chimera between proflavine-sensitive Kir1.1 and -insensitive Kir4.1 revealed that the extracellular portion of Kir1.1 is crucial for the recognition of the proflavine derivative acrinol. In conclusion, preferential blockage of Kir-channel family members by proflavine derivatives is based on multiple modes of action. This raises the possibility of designing subunit-specific inhibitors.
Collapse
Affiliation(s)
- Atsushi Inanobe
- Department of Pharmacology, Graduate School of Medicine (A.I., H.I., Y.K.), and Center for Advanced Medical Engineering and Informatics (A.I., Y.K.), Osaka University, Suita, Osaka, Japan
| | - Hideaki Itamochi
- Department of Pharmacology, Graduate School of Medicine (A.I., H.I., Y.K.), and Center for Advanced Medical Engineering and Informatics (A.I., Y.K.), Osaka University, Suita, Osaka, Japan
| | - Yoshihisa Kurachi
- Department of Pharmacology, Graduate School of Medicine (A.I., H.I., Y.K.), and Center for Advanced Medical Engineering and Informatics (A.I., Y.K.), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
17
|
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
|