1
|
Henderson SW, Nourmohammadi S, Ramesh SA, Yool AJ. Aquaporin ion conductance properties defined by membrane environment, protein structure, and cell physiology. Biophys Rev 2022; 14:181-198. [PMID: 35340612 PMCID: PMC8921385 DOI: 10.1007/s12551-021-00925-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/09/2021] [Indexed: 01/13/2023] Open
Abstract
Aquaporins (AQPs) are multifunctional transmembrane channel proteins permeable to water and an expanding array of solutes. AQP-mediated ion channel activity was first observed when purified AQP0 from bovine lens was incorporated into lipid bilayers. Electrophysiological properties of ion-conducting AQPs since discovered in plants, invertebrates, and mammals have been assessed using native, reconstituted, and heterologously expressed channels. Accumulating evidence is defining amino acid residues that govern differential solute permeability through intrasubunit and central pores of AQP tetramers. Rings of charged and hydrophobic residues around pores influence AQP selectivity, and are candidates for further work to define motifs that distinguish ion conduction capability, versus strict water and glycerol permeability. Similarities between AQP ion channels thus far include large single channel conductances and long open times, but differences in ionic selectivity, permeability to divalent cations, and mechanisms of gating (e.g., by voltage, pH, and cyclic nucleotides) are unique to subtypes. Effects of lipid environments in modulating parameters such as single channel amplitude could explain in part the variations in AQP ion channel properties observed across preparations. Physiological roles of the ion-conducting AQP classes span diverse processes including regulation of cell motility, organellar pH, neural development, signaling, and nutrient acquisition. Advances in computational methods can generate testable predictions of AQP structure-function relationships, which combined with innovative high-throughput assays could revolutionize the field in defining essential properties of ion-conducting AQPs, discovering new AQP ion channels, and understanding the effects of AQP interactions with proteins, signaling cascades, and membrane lipids.
Collapse
Affiliation(s)
- Sam W. Henderson
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005 Australia
| | | | - Sunita A. Ramesh
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042 Australia
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005 Australia
| |
Collapse
|
2
|
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: 17] [Impact Index Per Article: 5.7] [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
|
3
|
Bachmann M, Li W, Edwards MJ, Ahmad SA, Patel S, Szabo I, Gulbins E. Voltage-Gated Potassium Channels as Regulators of Cell Death. Front Cell Dev Biol 2020; 8:611853. [PMID: 33381507 PMCID: PMC7767978 DOI: 10.3389/fcell.2020.611853] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Ion channels allow the flux of specific ions across biological membranes, thereby determining ion homeostasis within the cells. Voltage-gated potassium-selective ion channels crucially contribute to the setting of the plasma membrane potential, to volume regulation and to the physiologically relevant modulation of intracellular potassium concentration. In turn, these factors affect cell cycle progression, proliferation and apoptosis. The present review summarizes our current knowledge about the involvement of various voltage-gated channels of the Kv family in the above processes and discusses the possibility of their pharmacological targeting in the context of cancer with special emphasis on Kv1.1, Kv1.3, Kv1.5, Kv2.1, Kv10.1, and Kv11.1.
Collapse
Affiliation(s)
- Magdalena Bachmann
- Department of Biology, University of Padova, Padua, Italy.,Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Weiwei Li
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Michael J Edwards
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Syed A Ahmad
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Sameer Patel
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy.,Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padua, Italy
| | - Erich Gulbins
- Department of Surgery, Medical School, University of Cincinnati, Cincinnati, OH, United States.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
4
|
Walsh KB. Screening Technologies for Inward Rectifier Potassium Channels: Discovery of New Blockers and Activators. SLAS DISCOVERY 2020; 25:420-433. [PMID: 32292089 DOI: 10.1177/2472555220905558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
K+ channels play a critical role in maintaining the normal electrical activity of excitable cells by setting the cell resting membrane potential and by determining the shape and duration of the action potential. In nonexcitable cells, K+ channels establish electrochemical gradients necessary for maintaining salt and volume homeostasis of body fluids. Inward rectifier K+ (Kir) channels typically conduct larger inward currents than outward currents, resulting in an inwardly rectifying current versus voltage relationship. This property of inward rectification results from the voltage-dependent block of the channels by intracellular polyvalent cations and makes these channels uniquely designed for maintaining the resting potential near the K+ equilibrium potential (EK). The Kir family of channels consist of seven subfamilies of channels (Kir1.x through Kir7.x) that include the classic inward rectifier (Kir2.x) channel, the G-protein-gated inward rectifier K+ (GIRK) (Kir3.x), and the adenosine triphosphate (ATP)-sensitive (KATP) (Kir 6.x) channels as well as the renal Kir1.1 (ROMK), Kir4.1, and Kir7.1 channels. These channels not only function to regulate electrical/electrolyte transport activity, but also serve as effector molecules for G-protein-coupled receptors (GPCRs) and as molecular sensors for cell metabolism. Of significance, Kir channels represent promising pharmacological targets for treating a number of clinical conditions, including cardiac arrhythmias, anxiety, chronic pain, and hypertension. This review provides a brief background on the structure, function, and pharmacology of Kir channels and then focuses on describing and evaluating current high-throughput screening (HTS) technologies, such as membrane potential-sensitive fluorescent dye assays, ion flux measurements, and automated patch clamp systems used for Kir channel drug discovery.
Collapse
Affiliation(s)
- Kenneth B Walsh
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
5
|
Xin J, Wan Mahtar WNA, Siah PC, Miswan N, Khoo BY. Potential use of Pichia pastoris strain SMD1168H expressing DNA topoisomerase I in the screening of potential anti‑breast cancer agents. Mol Med Rep 2019; 19:5368-5376. [PMID: 31059050 PMCID: PMC6522884 DOI: 10.3892/mmr.2019.10201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 02/28/2019] [Indexed: 11/05/2022] Open
Abstract
Cancer chemotherapy possesses high toxicity, particularly when a higher concentration of drugs is administered to patients. Therefore, searching for more effective compounds to reduce the toxicity of treatments, while still producing similar effects as current chemotherapy regimens, is required. Currently, the search for potential anticancer agents involves a random, inaccurate process with strategic deficits and a lack of specific targets. For this reason, the initial in vitro high‑throughput steps in the screening process should be reviewed for rapid identification of the compounds that may serve as anticancer agents. The present study aimed to investigate the potential use of the Pichia pastoris strain SMD1168H expressing DNA topoisomerase I (SMD1168H‑TOPOI) in a yeast‑based assay for screening potential anticancer agents. The cell density that indicated the growth of the recombinant yeast without treatment was first measured by spectrophotometry. Subsequently, the effects of glutamate (agonist) and camptothecin (antagonist) on the recombinant yeast cell density were investigated using the same approach, and finally, the effect of camptothecin on various cell lines was determined and compared with its effect on recombinant yeast. The current study demonstrated that growth was enhanced in SMD1168H‑TOPOI as compared with that in SMD1168H. Glutamate also enhanced the growth of the SMD1168H; however, the growth effect was not enhanced in SMD1168H‑TOPOI treated with glutamate. By contrast, camptothecin caused only lower cell density and growth throughout the treatment of SMD1168H‑TOPOI. The findings of the current study indicated that SMD1168H‑TOPOI has similar characteristics to MDA‑MB‑231 cells; therefore, it can be used in a yeast‑based assay to screen for more effective compounds that may inhibit the growth of highly metastatic breast cancer cells.
Collapse
Affiliation(s)
- Jian Xin
- Department of General Surgery, Taizhou First People's Hospital in Zhejiang Province, Taizhou, Zhejiang 318020, P.R. China
| | - Wan Nor Azlin Wan Mahtar
- Department of Microbiology, School of Biological Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Poh Chiew Siah
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Noorizan Miswan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Boon Yin Khoo
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| |
Collapse
|
6
|
Locascio A, Andrés-Colás N, Mulet JM, Yenush L. Saccharomyces cerevisiae as a Tool to Investigate Plant Potassium and Sodium Transporters. Int J Mol Sci 2019; 20:E2133. [PMID: 31052176 PMCID: PMC6539216 DOI: 10.3390/ijms20092133] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Sodium and potassium are two alkali cations abundant in the biosphere. Potassium is essential for plants and its concentration must be maintained at approximately 150 mM in the plant cell cytoplasm including under circumstances where its concentration is much lower in soil. On the other hand, sodium must be extruded from the plant or accumulated either in the vacuole or in specific plant structures. Maintaining a high intracellular K+/Na+ ratio under adverse environmental conditions or in the presence of salt is essential to maintain cellular homeostasis and to avoid toxicity. The baker's yeast, Saccharomyces cerevisiae, has been used to identify and characterize participants in potassium and sodium homeostasis in plants for many years. Its utility resides in the fact that the electric gradient across the membrane and the vacuoles is similar to plants. Most plant proteins can be expressed in yeast and are functional in this unicellular model system, which allows for productive structure-function studies for ion transporting proteins. Moreover, yeast can also be used as a high-throughput platform for the identification of genes that confer stress tolerance and for the study of protein-protein interactions. In this review, we summarize advances regarding potassium and sodium transport that have been discovered using the yeast model system, the state-of-the-art of the available techniques and the future directions and opportunities in this field.
Collapse
Affiliation(s)
- Antonella Locascio
- Instituto de Biología Molecular y Celular de Plantas, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, 46022 Valencia, Spain.
| | - Nuria Andrés-Colás
- Instituto de Biología Molecular y Celular de Plantas, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, 46022 Valencia, Spain.
| | - José Miguel Mulet
- Instituto de Biología Molecular y Celular de Plantas, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, 46022 Valencia, Spain.
| | - Lynne Yenush
- Instituto de Biología Molecular y Celular de Plantas, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, 46022 Valencia, Spain.
| |
Collapse
|
7
|
Sanson C, Schombert B, Filoche-Rommé B, Partiseti M, Bohme GA. Electrophysiological and Pharmacological Characterization of Human Inwardly Rectifying K ir2.1 Channels on an Automated Patch-Clamp Platform. Assay Drug Dev Technol 2019; 17:89-99. [PMID: 30835490 PMCID: PMC6479253 DOI: 10.1089/adt.2018.882] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inwardly rectifying IK1 potassium currents of the heart control the resting membrane potential of ventricular cardiomyocytes during diastole and contribute to their repolarization after each action potential. Mutations in the gene encoding Kir2.1 channels, which primarily conduct ventricular IK1, are associated with inheritable forms of arrhythmias and sudden cardiac death. Therefore, potential iatrogenic inhibition of Kir2.1-mediated IK1 currents is a cardiosafety concern during new drug discovery and development. Kir2.1 channels are part of the panel of cardiac ion channels currently considered for refined early compound risk assessment within the Comprehensive in vitro Proarrhythmia Assay initiative. In this study, we have validated a cell-based assay allowing functional quantification of Kir2.1 inhibitors using whole-cell recordings of Chinese hamster ovary cells stably expressing human Kir2.1 channels. We reproduced key electrophysiological and pharmacological features known for native IK1, including current enhancement by external potassium and voltage- and concentration-dependent blockade by external barium. Furthermore, the Kir inhibitors ML133, PA-6, and chloroquine, as well as the multichannel inhibitors chloroethylclonidine, chlorpromazine, SKF-96365, and the class III antiarrhythmic agent terikalant demonstrated slowly developing inhibitory activity in the low micromolar range. The robustness of this assay authorizes medium throughput screening for cardiosafety purposes and could help to enrich the currently limited Kir2.1 pharmacology.
Collapse
Affiliation(s)
- Camille Sanson
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Brigitte Schombert
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Bruno Filoche-Rommé
- 2 Integrated Drug Discovery, Medicinal Chemistry, Sanofi Research and Development, Vitry-sur-Seine, France
| | - Michel Partiseti
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| | - G Andrees Bohme
- 1 Integrated Drug Discovery, High-Content Biology, Sanofi Research and Development, Vitry-sur-Seine, France
| |
Collapse
|
8
|
Denny PW. Yeast: bridging the gap between phenotypic and biochemical assays for high-throughput screening. Expert Opin Drug Discov 2018; 13:1153-1160. [DOI: 10.1080/17460441.2018.1534826] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Paul W. Denny
- Department of Biosciences and Centre for Global Infectious Disease, Durham University, Durham, UK
| |
Collapse
|
9
|
Mackie TD, Brodsky JL. Investigating Potassium Channels in Budding Yeast: A Genetic Sandbox. Genetics 2018; 209:637-650. [PMID: 29967058 PMCID: PMC6028241 DOI: 10.1534/genetics.118.301026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/15/2018] [Indexed: 12/26/2022] Open
Abstract
Like all species, the model eukaryote Saccharomyces cerevisiae, or Bakers' yeast, concentrates potassium in the cytosol as an electrogenic osmolyte and enzyme cofactor. Yeast are capable of robust growth on a wide variety of potassium concentrations, ranging from 10 µM to 2.5 M, due to the presence of a high-affinity potassium uptake system and a battery of cation exchange transporters. Genetic perturbation of either of these systems retards yeast growth on low or high potassium, respectively. However, these potassium-sensitized yeast are a powerful genetic tool, which has been leveraged for diverse studies. Notably, the potassium-sensitive cells can be transformed with plasmids encoding potassium channels from bacteria, plants, or mammals, and subsequent changes in growth rate have been found to correlate with the activity of the introduced potassium channel. Discoveries arising from the use of this assay over the past three decades have increased our understanding of the structure-function relationships of various potassium channels, the mechanisms underlying the regulation of potassium channel function and trafficking, and the chemical basis of potassium channel modulation. In this article, we provide an overview of the major genetic tools used to study potassium channels in S. cerevisiae, a survey of seminal studies utilizing these tools, and a prospective for the future use of this elegant genetic approach.
Collapse
Affiliation(s)
- Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pennsylvania 15260
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pennsylvania 15260
| |
Collapse
|
10
|
O'Donnell BM, Mackie TD, Subramanya AR, Brodsky JL. Endoplasmic reticulum-associated degradation of the renal potassium channel, ROMK, leads to type II Bartter syndrome. J Biol Chem 2017. [PMID: 28630040 DOI: 10.1074/jbc.m117.786376] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type II Bartter syndrome is caused by mutations in the renal outer medullary potassium (ROMK) channel, but the molecular mechanisms underlying this disease are poorly defined. To rapidly screen for ROMK function, we developed a yeast expression system and discovered that yeast cells lacking endogenous potassium channels could be rescued by WT ROMK but not by ROMK proteins containing any one of four Bartter mutations. We also found that the mutant proteins were significantly less stable than WT ROMK. However, their degradation was slowed in the presence of a proteasome inhibitor or when yeast cells contained mutations in the CDC48 or SSA1 gene, which is required for endoplasmic reticulum (ER)-associated degradation (ERAD). Consistent with these data, sucrose gradient centrifugation and indirect immunofluorescence microscopy indicated that most ROMK protein was ER-localized. To translate these findings to a more relevant cell type, we measured the stabilities of WT ROMK and the ROMK Bartter mutants in HEK293 cells. As in yeast, the Bartter mutant proteins were less stable than the WT protein, and their degradation was slowed in the presence of a proteasome inhibitor. Finally, we discovered that low-temperature incubation increased the steady-state levels of a Bartter mutant, suggesting that the disease-causing mutation traps the protein in a folding-deficient conformation. These findings indicate that the underlying pathology for at least a subset of patients with type II Bartter syndrome is linked to the ERAD pathway and that future therapeutic strategies should focus on correcting deficiencies in ROMK folding.
Collapse
Affiliation(s)
- Brighid M O'Donnell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260; Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| |
Collapse
|
11
|
Ren S, Pang C, Li J, Huang Y, Zhang S, Zhan Y, An H. Styrax blocks inward and outward current of Kir2.1 channel. Channels (Austin) 2017; 11:46-54. [PMID: 27540685 DOI: 10.1080/19336950.2016.1207022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Kir2.1 plays key roles in setting rest membrane potential and modulation of cell excitability. Mutations of Kir2.1, such as D172N or E299V, inducing gain-of-function, can cause type3 short QT syndrome (SQT3) due to the enlarged outward currents. So far, there is no clinical drug target to block the currents of Kir2.1. Here, we identified a novel blocker of Kir2.1, styrax, which is a kind of natural compound selected from traditional Chinese medicine. Our data show that styrax can abolish the inward and outward currents of Kir2.1. The IC50 of styrax for WT, D172N and E299V are 0.0113 ± 0.00075, 0.0204 ± 0.0048 and 0.0122 ± 0.0012 (in volume), respectively. The results indicate that styrax can serve as a novel blocker for Kir2.1.
Collapse
Affiliation(s)
- Shuxi Ren
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| | - Chunli Pang
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| | - Junwei Li
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| | - Yayue Huang
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| | - Suhua Zhang
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| | - Yong Zhan
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| | - Hailong An
- a Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology , Tianjin , China
| |
Collapse
|
12
|
Stas JI, Bocksteins E, Jensen CS, Schmitt N, Snyders DJ. The anticonvulsant retigabine suppresses neuronal K V2-mediated currents. Sci Rep 2016; 6:35080. [PMID: 27734968 PMCID: PMC5062084 DOI: 10.1038/srep35080] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/20/2016] [Indexed: 11/09/2022] Open
Abstract
Enhancement of neuronal M-currents, generated through KV7.2-KV7.5 channels, has gained much interest for its potential in developing treatments for hyperexcitability-related disorders such as epilepsy. Retigabine, a KV7 channel opener, has proven to be an effective anticonvulsant and has recently also gained attention due to its neuroprotective properties. In the present study, we found that the auxiliary KCNE2 subunit reduced the KV7.2-KV7.3 retigabine sensitivity approximately 5-fold. In addition, using both mammalian expression systems and cultured hippocampal neurons we determined that low μM retigabine concentrations had ‘off-target’ effects on KV2.1 channels which have recently been implicated in apoptosis. Clinical retigabine concentrations (0.3–3 μM) inhibited KV2.1 channel function upon prolonged exposure. The suppression of the KV2.1 conductance was only partially reversible. Our results identified KV2.1 as a new molecular target for retigabine, thus giving a potential explanation for retigabine’s neuroprotective properties.
Collapse
Affiliation(s)
- Jeroen I Stas
- Laboratory for Molecular Biophysics, Physiology and Pharmacology, Department of Biomedical Sciences, University of Antwerp, CDE, Universiteitsplein 1, 2610 Antwerp, Belgium.,Ion Channel Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Elke Bocksteins
- Laboratory for Molecular Biophysics, Physiology and Pharmacology, Department of Biomedical Sciences, University of Antwerp, CDE, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Camilla S Jensen
- Ion Channel Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Nicole Schmitt
- Ion Channel Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Dirk J Snyders
- Laboratory for Molecular Biophysics, Physiology and Pharmacology, Department of Biomedical Sciences, University of Antwerp, CDE, Universiteitsplein 1, 2610 Antwerp, Belgium
| |
Collapse
|
13
|
Isolation of proflavine as a blocker of G protein-gated inward rectifier potassium channels by a cell growth-based screening system. Neuropharmacology 2016; 109:18-28. [PMID: 27236080 DOI: 10.1016/j.neuropharm.2016.05.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/11/2016] [Accepted: 05/24/2016] [Indexed: 01/29/2023]
Abstract
The overexpression of Kir3.2, a subunit of the G protein-gated inwardly rectifying K(+) channel, is implicated in some of the neurological phenotypes of Down syndrome (DS). Chemical compounds that block Kir3.2 are expected to improve the symptoms of DS. The purpose of this study is to develop a cell-based screening system to identify Kir3.2 blockers and then investigate the mode of action of the blocker. Chemical screening was carried out using a K(+) transporter-deficient yeast strain that expressed a constitutively active Kir3.2 mutant. The mode of action of an effective blocker was electrophysiologically analyzed using Kir channels expressed in Xenopus oocytes. Proflavine was identified to inhibit the growth of Kir3.2-transformant cells and Kir3.2 activity in a concentration-dependent manner. The current inhibition was strong when membrane potentials (Vm) was above equilibrium potential of K(+) (EK). When Vm was below EK, the blockage apparently depended on the difference between Vm and [K(+)]. Furthermore, the inhibition became stronger by lowering extracellular [K(+)]. These results indicated that the yeast strain serves as a screening system to isolate Kir3.2 blockers and proflavine is a prototype of a pore blocker of Kir3.2.
Collapse
|
14
|
Zhou TT, Quan LL, Chen LP, Du T, Sun KX, Zhang JC, Yu L, Li Y, Wan P, Chen LL, Jiang BH, Hu LH, Chen J, Shen X. SP6616 as a new Kv2.1 channel inhibitor efficiently promotes β-cell survival involving both PKC/Erk1/2 and CaM/PI3K/Akt signaling pathways. Cell Death Dis 2016; 7:e2216. [PMID: 27148689 PMCID: PMC4917657 DOI: 10.1038/cddis.2016.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/31/2022]
Abstract
Kv2.1 as a voltage-gated potassium (Kv) channel subunit has a pivotal role in the regulation of glucose-stimulated insulin secretion (GSIS) and pancreatic β-cell apoptosis, and is believed to be a promising target for anti-diabetic drug discovery, although the mechanism underlying the Kv2.1-mediated β-cell apoptosis is obscure. Here, the small molecular compound, ethyl 5-(3-ethoxy-4-methoxyphenyl)-2-(4-hydroxy-3-methoxybenzylidene)-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyrimidine-6-carboxylate (SP6616) was discovered to be a new Kv2.1 inhibitor. It was effective in both promoting GSIS and protecting β cells from apoptosis. Evaluation of SP6616 on either high-fat diet combined with streptozocin-induced type 2 diabetic mice or db/db mice further verified its efficacy in the amelioration of β-cell dysfunction and glucose homeostasis. SP6616 treatment efficiently increased serum insulin level, restored β-cell mass, decreased fasting blood glucose and glycated hemoglobin levels, and improved oral glucose tolerance. Mechanism study indicated that the promotion of SP6616 on β-cell survival was tightly linked to its regulation against both protein kinases C (PKC)/extracellular-regulated protein kinases 1/2 (Erk1/2) and calmodulin(CaM)/phosphatidylinositol 3-kinase(PI3K)/serine/threonine-specific protein kinase (Akt) signaling pathways. To our knowledge, this may be the first report on the underlying pathway responsible for the Kv2.1-mediated β-cell protection. In addition, our study has also highlighted the potential of SP6616 in the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- T T Zhou
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - L L Quan
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, China
| | - L P Chen
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - T Du
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - K X Sun
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - J C Zhang
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, China
| | - L Yu
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Y Li
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - P Wan
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, China
| | - L L Chen
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - B H Jiang
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - L H Hu
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - J Chen
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - X Shen
- CAS Key Laboratory of Receptor Research, 3th Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Yenush L. Potassium and Sodium Transport in Yeast. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 892:187-228. [DOI: 10.1007/978-3-319-25304-6_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
16
|
Okada M, Corzo G, Romero-Perez GA, Coronas F, Matsuda H, Possani LD. A pore forming peptide from spider Lachesana sp. venom induced neuronal depolarization and pain. Biochim Biophys Acta Gen Subj 2015; 1850:657-66. [DOI: 10.1016/j.bbagen.2014.11.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 11/12/2014] [Accepted: 11/28/2014] [Indexed: 10/24/2022]
|
17
|
Bagriantsev SN, Chatelain FC, Clark KA, Alagem N, Reuveny E, Minor DL. Tethered protein display identifies a novel Kir3.2 (GIRK2) regulator from protein scaffold libraries. ACS Chem Neurosci 2014; 5:812-22. [PMID: 25028803 PMCID: PMC4176385 DOI: 10.1021/cn5000698] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
![]()
Use of randomized peptide libraries
to evolve molecules with new
functions provides a means for developing novel regulators of protein
activity. Despite the demonstrated power of such approaches for soluble
targets, application of this strategy to membrane systems, such as
ion channels, remains challenging. Here, we have combined libraries
of a tethered protein scaffold with functional selection in yeast
to develop a novel activator of the G-protein-coupled mammalian inwardly
rectifying potassium channel Kir3.2 (GIRK2). We show that the novel
regulator, denoted N5, increases Kir3.2 (GIRK2) basal activity by
inhibiting clearance of the channel from the cellular surface rather
than affecting the core biophysical properties of the channel. These
studies establish the tethered protein display strategy as a means
to create new channel modulators and highlight the power of approaches
that couple randomized libraries with direct selections for functional
effects. Our results further underscore the possibility for the development
of modulators that influence channel function by altering cell surface
expression densities rather than by direct action on channel biophysical
parameters. The use of tethered library selection strategies coupled
with functional selection bypasses the need for a purified target
and is likely to be applicable to a range of membrane protein systems.
Collapse
Affiliation(s)
| | | | | | - Noga Alagem
- Department
of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eitan Reuveny
- Department
of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Daniel L. Minor
- Physical
Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
18
|
Denny PW, Steel PG. Yeast as a potential vehicle for neglected tropical disease drug discovery. ACTA ACUST UNITED AC 2014; 20:56-63. [PMID: 25121554 DOI: 10.1177/1087057114546552] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High-throughput screening (HTS) efforts for neglected tropical disease (NTD) drug discovery have recently received increased attention because several initiatives have begun to attempt to reduce the deficit in new and clinically acceptable therapies for this spectrum of infectious diseases. HTS primarily uses two basic approaches, cell-based and in vitro target-directed screening. Both of these approaches have problems; for example, cell-based screening does not reveal the target or targets that are hit, whereas in vitro methodologies lack a cellular context. Furthermore, both can be technically challenging, expensive, and difficult to miniaturize for ultra-HTS [(u)HTS]. The application of yeast-based systems may overcome some of these problems and offer a cost-effective platform for target-directed screening within a eukaryotic cell context. Here, we review the advantages and limitations of the technologies that may be used in yeast cell-based, target-directed screening protocols, and we discuss how these are beginning to be used in NTD drug discovery.
Collapse
Affiliation(s)
- P W Denny
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham, UK School of Medicine, Pharmacy and Health, Durham University, Durham, UK
| | - P G Steel
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham, UK
| |
Collapse
|
19
|
Mulet JM, Llopis-Torregrosa V, Primo C, Marqués MC, Yenush L. Endocytic regulation of alkali metal transport proteins in mammals, yeast and plants. Curr Genet 2013; 59:207-30. [PMID: 23974285 DOI: 10.1007/s00294-013-0401-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/24/2013] [Accepted: 07/29/2013] [Indexed: 12/30/2022]
Abstract
The relative concentrations of ions and solutes inside cells are actively maintained by several classes of transport proteins, in many cases against their concentration gradient. These transport processes, which consume a large portion of cellular energy, must be constantly regulated. Many structurally distinct families of channels, carriers, and pumps have been characterized in considerable detail during the past decades and defects in the function of some of these proteins have been linked to a growing list of human diseases. The dynamic regulation of the transport proteins present at the cell surface is vital for both normal cellular function and for the successful adaptation to changing environments. The composition of proteins present at the cell surface is controlled on both the transcriptional and post-translational level. Post-translational regulation involves highly conserved mechanisms of phosphorylation- and ubiquitylation-dependent signal transduction routes used to modify the cohort of receptors and transport proteins present under any given circumstances. In this review, we will summarize what is currently known about one facet of this regulatory process: the endocytic regulation of alkali metal transport proteins. The physiological relevance, major contributors, parallels and missing pieces of the puzzle in mammals, yeast and plants will be discussed.
Collapse
Affiliation(s)
- José Miguel Mulet
- Instituto de Biología Molecular y Celular de Plantas (IBMCP), Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avd. de los Naranjos s/n, 46022, Valencia, Spain
| | | | | | | | | |
Collapse
|
20
|
Bagriantsev SN, Ang KH, Gallardo-Godoy A, Clark KA, Arkin MR, Renslo AR, Minor DL. A high-throughput functional screen identifies small molecule regulators of temperature- and mechano-sensitive K2P channels. ACS Chem Biol 2013; 8:1841-51. [PMID: 23738709 PMCID: PMC3747594 DOI: 10.1021/cb400289x] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
K2P (KCNK) potassium channels
generate “leak”
potassium currents that strongly influence cellular excitability and
contribute to pain, somatosensation, anesthesia, and mood. Despite
their physiological importance, K2Ps lack specific pharmacology.
Addressing this issue has been complicated by the challenges that
the leak nature of K2P currents poses for electrophysiology-based
high-throughput screening strategies. Here, we present a yeast-based
high-throughput screening assay that avoids this problem. Using a
simple growth-based functional readout, we screened a library of 106,281
small molecules and identified two new inhibitors and three new activators
of the mammalian K2P channel K2P2.1 (KCNK2, TREK-1). By combining biophysical, structure–activity,
and mechanistic analysis, we developed a dihydroacridine analogue,
ML67-33, that acts as a low micromolar, selective activator of temperature-
and mechano-sensitive K2P channels. Biophysical studies
show that ML67-33 reversibly increases channel currents by activating
the extracellular selectivity filter-based C-type gate that forms
the core gating apparatus on which a variety of diverse modulatory
inputs converge. The new K2P modulators presented here,
together with the yeast-based assay, should enable both mechanistic
and physiological studies of K2P activity and facilitate
the discovery and development of other K2P small molecule
modulators.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniel L. Minor
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
94720, United States
| |
Collapse
|
21
|
Potential use of potassium efflux-deficient yeast for studying trafficking signals and potassium channel functions. FEBS Open Bio 2013; 3:196-203. [PMID: 23772394 PMCID: PMC3668539 DOI: 10.1016/j.fob.2013.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/02/2013] [Accepted: 04/11/2013] [Indexed: 11/22/2022] Open
Abstract
The activity of potassium (K(+)) channels critically depends on their density on the cell surface membrane, which is regulated by dynamic protein-protein interactions that often involve distinct trafficking signals on the cargo proteins. In this paper we explored the possibility of utilizing the Saccharomyces cerevisiae strain B31 for identification of the signal motifs that regulate surface expression of membrane proteins and for studying structure-function relationships of K(+) channels. B31 cells lack the K(+) efflux system and were reported to show overloaded K(+)-mediated growth inhibition in high K(+) media upon heterologous expression of a mammalian inwardly rectifying K(+) channel (Kir2.1). We show that while the expression of wild-type Kir2.1 channel inhibits the growth of B31 cells in high K(+) media, the human disease-causing mutations of Kir2.1 that abolish K(+) conduction (V302M) or surface trafficking (Δ314/315) fully restores the growth. The expression of two-pore-domain K(+) channel KCNK3 or KCNK9 also inhibited the growth of B31 in high K(+) media while C-terminal mutations that reduce their 14-3-3 protein-dependent cell surface trafficking restored the growth of B31. Finally, the expression of Kir2.1 channels that were C-terminally fused with known sequence motifs including ER retention/retrieval signals and an endocytosis signal allowed the growth of B31 in high K(+) media. These results demonstrate the potential of B31 yeast strain as a unique biological tool to screen the random peptide libraries for novel sequence signals that down-regulate surface expression of membrane proteins, as well as to systematically identify the structural determinants for cell surface trafficking and/or ion conductance of K(+) channels.
Collapse
|
22
|
The utility of yeast as a tool for cell-based, target-directed high-throughput screening. Parasitology 2013; 141:8-16. [PMID: 23611102 DOI: 10.1017/s0031182013000425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many Neglected Tropical Diseases (NTDs) have recently been subject of increased focus, particularly with relation to high-throughput screening (HTS) initiatives. These vital endeavours largely rely of two approaches, in vitro target-directed screening using biochemical assays or cell-based screening which takes no account of the target or targets being hit. Despite their successes both of these approaches have limitations; for example, the production of soluble protein and a lack of cellular context or the problems and expense of parasite cell culture. In addition, both can be challenging to miniaturize for ultra (u)HTS and expensive to utilize. Yeast-based systems offer a cost-effective approach to study and screen protein targets in a direct-directed manner within a eukaryotic cellular context. In this review, we examine the utility and limitations of yeast cell-based, target-directed screening. In particular we focus on the currently under-explored possibility of using such formats in uHTS screening campaigns for NTDs.
Collapse
|
23
|
Using yeast to study potassium channel function and interactions with small molecules. Methods Mol Biol 2013; 995:31-42. [PMID: 23494370 DOI: 10.1007/978-1-62703-345-9_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Analysis of ion channel mutants is a widely used approach for dissecting ion channel function and for characterizing the mechanisms of action of channel-directed modulators. Expression of functional potassium channels in potassium-uptake-deficient yeast together with genetic selection approaches offers an unbiased, high-throughput, activity-based readout that can rapidly identify large numbers of active ion channel mutants. Because of the assumption-free nature of the method, detailed biophysical analysis of the functional mutants from such selections can provide new and unexpected insights into both ion channel gating and ion channel modulator mechanisms. Here, we present detailed protocols for generation and identification of functional mutations in potassium channels using yeast selections in the potassium-uptake-deficient strain SGY1528. This approach is applicable for the analysis of structure-function relationships of potassium channels from a wide range of sources including viruses, bacteria, plants, and mammals and can be used as a facile way to probe the interactions between ion channels and small-molecule modulators.
Collapse
|
24
|
D'Onofrio PM, Koeberle PD. What can we learn about stroke from retinal ischemia models? Acta Pharmacol Sin 2013. [PMID: 23202803 DOI: 10.1038/aps.2012.165] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Retinal ischemia is a very useful model to study the impact of various cell death pathways, such as apoptosis and necrosis, in the ischemic retina. However, it is important to note that the retina is formed as an outpouching of the diencephalon and is part of the central nervous system. As such, the cell death pathways initiated in response to ischemic damage in the retina reflect those found in other areas of the central nervous system undergoing similar trauma. The retina is also more accessible than other areas of the central nervous system, thus making it a simpler model to work with and study. By utilizing the retinal model, we can greatly increase our knowledge of the cell death processes initiated by ischemia which lead to degeneration in the central nervous system. This paper examines work that has been done so far to characterize various aspects of cell death in the retinal ischemia model, such as various pathways which are activated, and the role neurotrophic factors, and discusses how these are relevant to the treatment of ischemic damage in both the retina and the greater central nervous system.
Collapse
|
25
|
McFerrin MB, Turner KL, Cuddapah VA, Sontheimer H. Differential role of IK and BK potassium channels as mediators of intrinsic and extrinsic apoptotic cell death. Am J Physiol Cell Physiol 2012; 303:C1070-8. [PMID: 22992678 DOI: 10.1152/ajpcell.00040.2012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An important event during apoptosis is regulated cell condensation known as apoptotic volume decrease (AVD). Ion channels have emerged as essential regulators of this process mediating the release of K(+) and Cl(-), which together with osmotically obliged water, results in the condensation of cell volume. Using a Grade IV human glioblastoma cell line, we examined the contribution of calcium-activated K(+) channels (K(Ca) channels) to AVD after the addition of either staurosporine (Stsp) or TNF-α-related apoptosis-inducing ligand (TRAIL) to activate the intrinsic or extrinsic pathway of apoptosis, respectively. We show that AVD can be inhibited in both pathways by high extracellular K(+) or the removal of calcium. However, BAPTA-AM was only able to inhibit Stsp-initiated AVD, whereas TRAIL-induced AVD was unaffected. Specific K(Ca) channel inhibitors revealed that Stsp-induced AVD was dependent on K(+) efflux through intermediate-conductance calcium-activated potassium (IK) channels, while TRAIL-induced AVD was mediated by large-conductance calcium-activated potassium (BK) channels. Fura-2 imaging demonstrated that Stsp induced a rapid and modest rise in calcium that was sustained over the course of AVD, while TRAIL produced no detectable rise in global intracellular calcium. Inhibition of IK channels with clotrimazole or 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole (TRAM-34) blocked downstream caspase-3 activation after Stsp addition, while paxilline, a specific BK channel inhibitor, had no effect. Treatment with ionomycin also induced an IK-dependent cell volume decrease. Together these results show that calcium is both necessary and sufficient to achieve volume decrease and that the two major pathways of apoptosis use unique calcium signaling to efflux K(+) through different K(Ca) channels.
Collapse
Affiliation(s)
- Michael B McFerrin
- Deptartment of Neurobiology and Center for Glial Biology in Medicine, Universtiy of Alabama at Birmingham, 1719 6th Ave. South, CIRC 410, Birmingham, AL, 35294, USA
| | | | | | | |
Collapse
|
26
|
Fujii M, Ohya S, Yamamura H, Imaizumi Y. Development of recombinant cell line co-expressing mutated Nav1.5, Kir2.1, and hERG for the safety assay of drug candidates. ACTA ACUST UNITED AC 2012; 17:773-84. [PMID: 22498908 DOI: 10.1177/1087057112442102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To provide a high-throughput screening method for human ether-a-go-go-gene-related gene (hERG) K(+) channel inhibition, a new recombinant cell line, in which single action potential (AP)-induced cell death was produced by gene transfection. Mutated human cardiac Na(+) channel Nav1.5 (IFM/Q3), which shows extremely slow inactivation, and wild-type inward rectifier K(+) channel, Kir2.1, were stably co-expressed in HEK293 cells (IFM/Q3+Kir2.1). In IFM/Q3+Kir2.1, application of single electrical stimulation (ES) elicited a long AP lasting more than 30 s and led cells to die by more than 70%, whereas HEK293 co-transfected with wild-type Nav1.5 and Kir2.1 fully survived. The additional expression of hERG K(+) channels in IFM/Q3+Kir2.1 shortened the duration of evoked AP and thereby markedly reduced the cell death. The treatment of the cells with hERG channel inhibitors such as nifekalant, E-4031, cisapride, terfenadine, and verapamil, recovered the prolonged AP and dose-dependently facilitated cell death upon ES. The EC(50) values to induce the cell death were 3 µM, 19 nM, 17 nM, 74 nM, and 3 µM, respectively, whereas 10 µM nifedipine did not induce cell death. Results indicate the high utility of this cell system for hERG K(+) channel safety assay.
Collapse
|
27
|
Hao S, Bao YM, An LJ, Cheng W, Zhao RG, Bi J, Wang HS, Sun CS, Liu JW, Jiang B. Effects of Resibufogenin and Cinobufagin on voltage-gated potassium channels in primary cultures of rat hippocampal neurons. Toxicol In Vitro 2011; 25:1644-53. [DOI: 10.1016/j.tiv.2011.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 05/29/2011] [Accepted: 07/04/2011] [Indexed: 10/17/2022]
|
28
|
Wang HR, Wu M, Yu H, Long S, Stevens A, Engers DW, Sackin H, Daniels JS, Dawson ES, Hopkins CR, Lindsley CW, Li M, McManus OB. Selective inhibition of the K(ir)2 family of inward rectifier potassium channels by a small molecule probe: the discovery, SAR, and pharmacological characterization of ML133. ACS Chem Biol 2011; 6:845-56. [PMID: 21615117 DOI: 10.1021/cb200146a] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The K(ir) inward rectifying potassium channels have a broad tissue distribution and are implicated in a variety of functional roles. At least seven classes (K(ir)1-K(ir)7) of structurally related inward rectifier potassium channels are known, and there are no selective small molecule tools to study their function. In an effort to develop selective K(ir)2.1 inhibitors, we performed a high-throughput screen (HTS) of more than 300,000 small molecules within the MLPCN for modulators of K(ir)2.1 function. Here we report one potent K(ir)2.1 inhibitor, ML133, which inhibits K(ir)2.1 with an IC(50) of 1.8 μM at pH 7.4 and 290 nM at pH 8.5 but exhibits little selectivity against other members of Kir2.x family channels. However, ML133 has no effect on K(ir)1.1 (IC(50) > 300 μM) and displays weak activity for K(ir)4.1 (76 μM) and K(ir)7.1 (33 μM), making ML133 the most selective small molecule inhibitor of the K(ir) family reported to date. Because of the high homology within the K(ir)2 family-the channels share a common design of a pore region flanked by two transmembrane domains-identification of site(s) critical for isoform specificity would be an important basis for future development of more specific and potent K(ir) inhibitors. Using chimeric channels between K(ir)2.1 and K(ir)1.1 and site-directed mutagenesis, we have identified D172 and I176 within M2 segment of K(ir)2.1 as molecular determinants critical for the potency of ML133 mediated inhibition. Double mutation of the corresponding residues of K(ir)1.1 to those of K(ir)2.1 (N171D and C175I) transplants ML133 inhibition to K(ir)1.1. Together, the combination of a potent, K(ir)2 family selective inhibitor and identification of molecular determinants for the specificity provides both a tool and a model system to enable further mechanistic studies of modulation of K(ir)2 inward rectifier potassium channels.
Collapse
Affiliation(s)
- Hao-Ran Wang
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| | - Meng Wu
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| | - Haibo Yu
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| | - Shunyou Long
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| | - Amy Stevens
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| | - Darren W. Engers
- Vanderbilt Specialized Chemistry Center, Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Henry Sackin
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, Ilinois 60064, United States
| | - J. Scott Daniels
- Vanderbilt Specialized Chemistry Center, Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Eric S. Dawson
- Vanderbilt Specialized Chemistry Center, Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Corey R. Hopkins
- Vanderbilt Specialized Chemistry Center, Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Vanderbilt Specialized Chemistry Center, Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Min Li
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| | - Owen B. McManus
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, School of Medicine, Johns Hopkins University, 733 North Broadway, Baltimore, Maryland 21205, United States
| |
Collapse
|
29
|
Small-molecule modulators of inward rectifier K+ channels: recent advances and future possibilities. Future Med Chem 2011; 2:757-74. [PMID: 20543968 DOI: 10.4155/fmc.10.179] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Inward rectifier potassium (Kir) channels have been postulated as therapeutic targets for several common disorders including hypertension, cardiac arrhythmias and pain. With few exceptions, however, the small-molecule pharmacology of this family is limited to nonselective cardiovascular and neurologic drugs with off-target activity toward inward rectifiers. Consequently, the actual therapeutic potential and 'drugability' of most Kir channels has not yet been determined experimentally. The purpose of this review is to provide a comprehensive summary of publicly disclosed Kir channel small-molecule modulators and highlight recent targeted drug-discovery efforts toward Kir1.1 and Kir2.1. The review concludes with a brief speculation on how the field of Kir channel pharmacology will develop over the coming years and a discussion of the increasingly important role academic laboratories will play in this progress.
Collapse
|
30
|
Kolb AR, Buck TM, Brodsky JL. Saccharomyces cerivisiae as a model system for kidney disease: what can yeast tell us about renal function? Am J Physiol Renal Physiol 2011; 301:F1-11. [PMID: 21490136 PMCID: PMC3129885 DOI: 10.1152/ajprenal.00141.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 04/11/2011] [Indexed: 01/18/2023] Open
Abstract
Ion channels, solute transporters, aquaporins, and factors required for signal transduction are vital for kidney function. Because mutations in these proteins or in associated regulatory factors can lead to disease, an investigation into their biogenesis, activities, and interplay with other proteins is essential. To this end, the yeast, Saccharomyces cerevisiae, represents a powerful experimental system. Proteins expressed in yeast include the following: 1) ion channels, including the epithelial sodium channel, members of the inward rectifying potassium channel family, and cystic fibrosis transmembrane conductance regulator; 2) plasma membrane transporters, such as the Na(+)-K(+)-ATPase, the Na(+)-phosphate cotransporter, and the Na(+)-H(+) ATPase; 3) aquaporins 1-4; and 4) proteins such as serum/glucocorticoid-induced kinase 1, phosphoinositide-dependent kinase 1, Rh glycoprotein kidney, and trehalase. The variety of proteins expressed and studied emphasizes the versatility of yeast, and, because of the many available tools in this organism, results can be obtained rapidly and economically. In most cases, data gathered using yeast have been substantiated in higher cell types. These attributes validate yeast as a model system to explore renal physiology and suggest that research initiated using this system may lead to novel therapeutics.
Collapse
Affiliation(s)
- Alexander R Kolb
- University of Pittsburgh, 4249 Fifth Ave., A320 Langley Hall, Pittsburgh, PA 15260, USA
| | | | | |
Collapse
|
31
|
Zou B, Yu H, Babcock JJ, Chanda P, Bader JS, McManus OB, Li M. Profiling diverse compounds by flux- and electrophysiology-based primary screens for inhibition of human Ether-à-go-go related gene potassium channels. Assay Drug Dev Technol 2011; 8:743-54. [PMID: 21158688 DOI: 10.1089/adt.2010.0339] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Compound effects on cloned human Ether-à-go-go related gene (hERG) potassium channels have been used to assess the potential cardiac safety liabilities of drug development candidate compounds. In addition to radioactive ligand displacement tests, two other common approaches are surrogate ion-based flux assays and electrophysiological recordings. The former has much higher throughput, whereas the latter measures directly the effects on ionic currents. Careful characterization in earlier reports has been performed to compare the relative effectiveness of these approaches for known hERG blockers, which often yielded good overall correlation. However, cases were reported showing significant and reproducible differences in potency and/or sensitivity by the two methods. This raises a question concerning the rationale and criteria on which an assay should be selected for evaluating unknown compounds. To provide a general basis for considering assays to profile large compound libraries for hERG activity, we have conducted parallel flux and electrophysiological analyses of 2,000 diverse compounds, representative of the 300,000 compound collection of NIH Molecular Library Small Molecular Repository (MLSMR). Our results indicate that at the conventional testing concentration 1.0 μM, the overlap between the two assays ranges from 32% to 50% depending on the hit selection criteria. There was a noticeable rate of false negatives by the thallium-based assay relative to electrophysiological recording, which may be greatly reduced under modified comparative conditions. As these statistical results identify a preferred method for cardiac safety profiling of unknown compounds, they suggest an efficient method combining flux and electrophysiological assays to rapidly profile hERG liabilities of large collection of naive compounds.
Collapse
Affiliation(s)
- Beiyan Zou
- Department of Neuroscience, High Throughput Biology Center, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Demirbas D, Ceyhan O, Wyman AR, Hoffman CS. A fission yeast-based platform for phosphodiesterase inhibitor HTSs and analyses of phosphodiesterase activity. Handb Exp Pharmacol 2011:135-49. [PMID: 21695638 DOI: 10.1007/978-3-642-17969-3_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fission yeast strains have been engineered so that their growth behavior reflects the activity of heterologous cyclic nucleotide phosphodiesterases (PDEs). These strains can be used in High-Throughput Screens (HTSs) for PDE inhibitors that possess "drug-like" characteristics, displaying activity in a growth stimulation assay over a 48-h period. Through three generations of development, a collection of strains expressing 10 of the 11 mammalian PDE families that is appropriate for small molecule inhibitor screening has been generated in our laboratory. Strains unable to synthesize cyclic nucleotides allow characterization of PDE activity in that the enzyme's potency is reflected in the amount of either cAMP or cGMP that must be added to the growth medium to stimulate cell growth. In the future, this system could be used to screen cDNA libraries for biological regulators of target PDEs and for the construction of strains that co-express PDEs and associated regulatory proteins to facilitate molecular and genetic studies of their functions and, in particular, to identify whether different PDE-partner protein complexes show distinct patterns of inhibitor sensitivity.
Collapse
Affiliation(s)
- Didem Demirbas
- Biology Department, Boston College, Higgins Hall 401B, Chestnut Hill, Boston, MA 02467, USA
| | | | | | | |
Collapse
|
33
|
Chatelain FC, Gazzarrini S, Fujiwara Y, Arrigoni C, Domigan C, Ferrara G, Pantoja C, Thiel G, Moroni A, Minor DL. Selection of inhibitor-resistant viral potassium channels identifies a selectivity filter site that affects barium and amantadine block. PLoS One 2009; 4:e7496. [PMID: 19834614 PMCID: PMC2759520 DOI: 10.1371/journal.pone.0007496] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Accepted: 09/23/2009] [Indexed: 12/02/2022] Open
Abstract
Background Understanding the interactions between ion channels and blockers remains an important goal that has implications for delineating the basic mechanisms of ion channel function and for the discovery and development of ion channel directed drugs. Methodology/Principal Findings We used genetic selection methods to probe the interaction of two ion channel blockers, barium and amantadine, with the miniature viral potassium channel Kcv. Selection for Kcv mutants that were resistant to either blocker identified a mutant bearing multiple changes that was resistant to both. Implementation of a PCR shuffling and backcrossing procedure uncovered that the blocker resistance could be attributed to a single change, T63S, at a position that is likely to form the binding site for the inner ion in the selectivity filter (site 4). A combination of electrophysiological and biochemical assays revealed a distinct difference in the ability of the mutant channel to interact with the blockers. Studies of the analogous mutation in the mammalian inward rectifier Kir2.1 show that the T→S mutation affects barium block as well as the stability of the conductive state. Comparison of the effects of similar barium resistant mutations in Kcv and Kir2.1 shows that neighboring amino acids in the Kcv selectivity filter affect blocker binding. Conclusions/Significance The data support the idea that permeant ions have an integral role in stabilizing potassium channel structure, suggest that both barium and amantadine act at a similar site, and demonstrate how genetic selections can be used to map blocker binding sites and reveal mechanistic features.
Collapse
Affiliation(s)
- Franck C. Chatelain
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Sabrina Gazzarrini
- Dipartimento di Biologia e Istituto di Biofisica del Consiglio Nazionale delle Ricerche, Università degli Studi di Milano, Milan, Italy
| | - Yuichiro Fujiwara
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Cristina Arrigoni
- Dipartimento di Biologia e Istituto di Biofisica del Consiglio Nazionale delle Ricerche, Università degli Studi di Milano, Milan, Italy
| | - Courtney Domigan
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Giuseppina Ferrara
- Dipartimento di Biologia e Istituto di Biofisica del Consiglio Nazionale delle Ricerche, Università degli Studi di Milano, Milan, Italy
| | - Carlos Pantoja
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | - Gerhard Thiel
- Technische Universität Darmstadt, Institute für Botanik, Darmstadt, Germany
| | - Anna Moroni
- Dipartimento di Biologia e Istituto di Biofisica del Consiglio Nazionale delle Ricerche, Università degli Studi di Milano, Milan, Italy
| | - Daniel L. Minor
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
- Department of California Institute for Quantitative Biomedical Research, University of California San Francisco, San Francisco, California, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Minor DL. Searching for interesting channels: pairing selection and molecular evolution methods to study ion channel structure and function. MOLECULAR BIOSYSTEMS 2009; 5:802-10. [PMID: 19603113 DOI: 10.1039/b901708a] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pairing of selection and screening methods with randomly mutated libraries can be an exceptionally powerful means for probing the functions of biological molecules and for developing novel regents from random libraries of peptides and oligonucleotides. The use of such approaches is beginning to permeate the ion channel field where they are being deployed to uncover fundamental aspects about ion channel structure and gating, small molecule-channel interactions, and the development of novel agents to control channel activity.
Collapse
Affiliation(s)
- Daniel L Minor
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, California Institute for Quantitative Biosciences, University of California, San Francisco, CA 94158-2330, USA.
| |
Collapse
|
35
|
Arylbenzazepines are potent modulators for the delayed rectifier K+ channel: a potential mechanism for their neuroprotective effects. PLoS One 2009; 4:e5811. [PMID: 19503734 PMCID: PMC2690691 DOI: 10.1371/journal.pone.0005811] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 05/06/2009] [Indexed: 11/19/2022] Open
Abstract
(+/-) SKF83959, like many other arylbenzazepines, elicits powerful neuroprotection in vitro and in vivo. The neuroprotective action of the compound was found to partially depend on its D(1)-like dopamine receptor agonistic activity. The precise mechanism for the (+/-) SKF83959-mediated neuroprotection remains elusive. We report here that (+/-) SKF83959 is a potent blocker for delayed rectifier K(+) channel. (+/-) SKF83959 inhibited the delayed rectifier K(+) current (I(K)) dose-dependently in rat hippocampal neurons. The IC(50) value for inhibition of I(K) was 41.9+/-2.3 microM (Hill coefficient = 1.81+/-0.13, n = 6), whereas that for inhibition of I(A) was 307.9+/-38.5 microM (Hill coefficient = 1.37+/-0.08, n = 6). Thus, (+/-) SKF83959 is 7.3-fold more potent in suppressing I(K) than I(A). Moreover, the inhibition of I(K) by (+/-) SKF83959 was voltage-dependent and not related to dopamine receptors. The rapidly onset of inhibition and recovery suggests that the inhibition resulted from a direct interaction of (+/-) SKF83959 with the K(+) channel. The intracellular application of (+/-) SKF83959 had no effects of on I(K), indicating that the compound most likely acts at the outer mouth of the pore of K(+) channel. We also tested the enantiomers of (+/-) SKF83959, R-(+) SKF83959 (MCL-201), and S-(-) SKF83959 (MCL-202), as well as SKF38393; all these compounds inhibited I(K). However, (+/-) SKF83959, at either 0.1 or 1 mM, exhibited the strongest inhibition on the currents among all tested drug. The present findings not only revealed a new potent blocker of I(K) , but also provided a novel mechanism for the neuroprotective action of arylbenzazepines such as (+/-) SKF83959.
Collapse
|
36
|
Zaks-Makhina E, Li H, Grishin A, Salvador-Recatala V, Levitan ES. Specific and slow inhibition of the kir2.1 K+ channel by gambogic acid. J Biol Chem 2009; 284:15432-8. [PMID: 19366693 DOI: 10.1074/jbc.m901586200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although Kir2.1 channels are important in the heart and other excitable cells, there are virtually no specific drugs for this K+ channel. In search of Kir2.1 modulators, we screened a library of 720 naturally occurring compounds using a yeast strain in which mammalian Kir2.1 enables growth at low [K+]. One of the identified compounds, gambogic acid (GA), potently (EC(50) < or = 100 nm) inhibited Kir2.1 channels in mammalian cells when applied chronically for 3 h. This potent and slow inhibition was not seen with Kv2.1, HERG or Kir1.1 channels. However, acutely applied GA acted as a weak (EC(50) = approximately 10 mum) non-selective K+ channel blocker. Intracellular delivery of GA via a patch pipette did not potentiate the acute effect of GA on Kir2.1, showing that slow uptake is not responsible for the delayed, potent effect. Immunoblots showed that total Kir2.1 protein expression was not altered by GA. Similarly, immunostaining of intact cells expressing Kir2.1 with an extracellular epitope tag demonstrated that GA does not affect Kir2.1 surface expression. However, the 3-h treatment with GA caused redistribution of Kir2.1 and Kv2.1 from the Triton X-100-insoluble to the Triton X-100-soluble membrane fraction. Thus, GA changes the K+ channel membrane microenvironment resulting in potent, specific, and slow acting inhibition of Kir2.1 channels.
Collapse
Affiliation(s)
- Elena Zaks-Makhina
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|
37
|
Legros C, Guette C, Martin-Eauclaire MF, Goyffon M, Tortajada J. Affinity capture using chimeric membrane proteins bound to magnetic beads for rapid ligand screening by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2009; 23:745-755. [PMID: 19204930 DOI: 10.1002/rcm.3939] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The rapid and specific detection of therapeutically important ligands in complex mixtures, that may bind to membrane proteins, remains challenging for many research laboratories and pharmaceutical industries. Through its use in the development of screening assays, mass spectrometry (MS) is currently experiencing a period of tremendous expansion. In the study presented here, we took advantage of the remarkable stability properties of a bacterial membrane protein, the KcsA K+ channel, produced in E. coli and purified as a tetrameric protein in the presence of a detergent. This membrane protein can subserve as a molecular template to display the pore-forming region of human K+ channels, which are considered as targets in the search for inhibitory ligands. The engineered chimeric proteins were linked to metal-bound magnetic beads, for the screening of complex peptide mixtures, such as that of scorpion venoms. The affinity-captured scorpion toxins were eluted prior to matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS), and to nano-electrospray ionization tandem mass QqTOF mass spectrometry (MS/MS) analysis. The de novo sequence of the toxins was deduced by combining the MS/MS fragmentation of the reduced form (up to the 33 first residues) and the trypsin digest peptides of the native toxins. This affinity-capture screening assay led to the isolation and characterization of potent and specific ligands of the human K+ channel, Kv1.3. The affinity-capture procedure is fast and reproducible. When linked to magnetic beads, the chimeric membrane protein can be re-used several times without losing any of its selectivity or specificity. This assay also benefits from the fact that it requires minimal amounts of animal venoms or complex mixtures, which can be expensive or difficult to procure.
Collapse
Affiliation(s)
- Christian Legros
- Laboratoire Analyse et Modélisation pour la Biologie et l'Environnement (LAMBE), Université d'Evry Val d'Essonne, CNRS UMR 8587, Bd F. Mitterrand, 91025 Evry, France.
| | | | | | | | | |
Collapse
|
38
|
Paynter JJ, Sarkies P, Andres-Enguix I, Tucker SJ. Genetic selection of activatory mutations in KcsA. Channels (Austin) 2008; 2:413-8. [PMID: 18797191 DOI: 10.4161/chan.2.6.6874] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The KcsA potassium channel from Streptomyces lividans is one of the most actively studied ion channels. However, there are still unresolved issues about its gating mechanism in vivo because the channel is only activated by highly acidic intracellular pH, meaning that it will be mostly inactive in its host environment. In this study we have used a genetic complementation assay of K+-auxotrophic E. coli (TK2420) and S. cerevisiae (SGY1528) to identify activatory or 'gain-of-function' mutations which allow functional activity of KcsA in the physiological environment of two markedly different expression systems. These mutations clustered at the helix-bundle-crossing in both TM1 and TM2 (residues H25, L105, A108, T112, W113, F114, E118 and Q119), and include residues previously implicated in the pH-gating mechanism. We discuss how these gain-of-function mutations may result in their activatory phenotype, the relative merits of the E. coli and S. cerevisiae genetic complementation approaches for the identification of gating mutations in prokaryotic K+ channels, and ways in which this assay may be improved for future use in screening protocols.
Collapse
Affiliation(s)
- Jennifer J Paynter
- Oxford Centre for Gene Function, Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
39
|
Song MK, Liu H, Jiang HL, Yue JM, Hu GY, Chen HZ. Discovery of talatisamine as a novel specific blocker for the delayed rectifier K+ channels in rat hippocampal neurons. Neuroscience 2008; 155:469-75. [DOI: 10.1016/j.neuroscience.2008.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 05/27/2008] [Accepted: 06/04/2008] [Indexed: 01/01/2023]
|
40
|
Jessica Chen M, Sepramaniam S, Armugam A, Shyan Choy M, Manikandan J, Melendez AJ, Jeyaseelan K, Sang Cheung N. Water and ion channels: crucial in the initiation and progression of apoptosis in central nervous system? Curr Neuropharmacol 2008; 6:102-16. [PMID: 19305791 PMCID: PMC2647147 DOI: 10.2174/157015908784533879] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 09/09/2007] [Accepted: 10/01/2007] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death (PCD), is a highly regulated and sophisticated cellular mechanism that commits cell to isolated death fate. PCD has been implicated in the pathogenesis of numerous neurodegenerative disorders. Countless molecular events underlie this phenomenon, with each playing a crucial role in death commitment. A precedent event, apoptotic volume decrease (AVD), is ubiquitously observed in various forms of PCD induced by different cellular insults. Under physiological conditions, cells when subjected to osmotic fluctuations will undergo regulatory volume increase/decrease (RVI/RVD) to achieve homeostatic balance with neurons in the brain being additionally protected by the blood-brain-barrier. However, during AVD following apoptotic trigger, cell undergoes anistonic shrinkage that involves the loss of water and ions, particularly monovalent ions e.g. K(+), Na(+) and Cl(-). It is worthwhile to concentrate on the molecular implications underlying the loss of these cellular components which posed to be significant and crucial in the successful propagation of the apoptotic signals. Microarray and real-time PCR analyses demonstrated several ion and water channel genes are regulated upon the onset of lactacystin (a proteosomal inhibitor)-mediated apoptosis. A time course study revealed that gene expressions of water and ion channels are being modulated just prior to apoptosis, some of which are aquaporin 4 and 9, potassium channels and chloride channels. In this review, we shall looked into the molecular protein machineries involved in the execution of AVD in the central nervous system (CNS), and focus on the significance of movements of each cellular component in affecting PCD commitment, thus provide some pharmacological advantages in the global apoptotic cell death.
Collapse
Affiliation(s)
- Minghui Jessica Chen
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Sugunavathi Sepramaniam
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Arunmozhiarasi Armugam
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Meng Shyan Choy
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jayapal Manikandan
- Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Alirio J Melendez
- Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Kandiah Jeyaseelan
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Nam Sang Cheung
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
41
|
Gao ZB, Chen XQ, Jiang HL, Liu H, Hu GY. Electrophysiological characterization of a novel Kv channel blocker N,N'-[oxybis(2,1-ethanediyloxy-2,1-ethanediyl) ]bis(4-methyl)-benzenesulfonamide found in virtual screening. Acta Pharmacol Sin 2008; 29:405-12. [PMID: 18358085 DOI: 10.1111/j.1745-7254.2008.00777.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
AIM N,No-[oxybis(2,1-ethanediyloxy-2,1-ethanediyl)]bis(4-methyl)- benzenesulfonamide (OMBSA) is a hit compound with potent voltage-gated K+ (Kv) channel-blocking activities that was found while searching the MDL Available Chemicals Directory with a virtual screening approach. In the present study, the blocking actions of OMBSA on Kv channels and relevant mechanisms were characterized. METHODS Whole-cell voltage-clamp recording was made in acutely dissociated hippocampal CA1 pyramidal neurons of newborn rats. RESULTS Superfusion of OMBSA reversibly inhibited both the delayed rectifier (I(K)) and fast transient K+ currents (I(A)) with IC50 values of 2.1+/-1.1 micromol/L and 27.8+/-1.5 micromol/L, respectively. The inhibition was voltage independent. OMBSA markedly accelerated the decay time course of IK, without a significant effect on that of I(A). OMBSA did not change the activation, steady-state inactivation of IK, and its recovery from inactivation, but the compound caused a significant hyperpolarizing shift of the voltage dependence of the steady-state inactivation of I(A) and slowed down its recovery from inactivation. Intracellular dialysis of OMBSA had no effect on both I(K) and I(A). CONCLUSION The results demonstrate that OMBSA blocks both I(K) and I(A) through binding to the outer mouth of the channel pore, as predicted by the molecular docking model used in the virtual screening. In addition, the compound differentially moderates the inactivation kinetics of the K+ channels through allosteric mechanisms.
Collapse
Affiliation(s)
- Zhao-bing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | | | | | | | | |
Collapse
|
42
|
Schwarzer S, Kolacna L, Lichtenberg-Fraté H, Sychrova H, Ludwig J. Functional expression of the voltage-gated neuronal mammalian potassium channel rat ether à go-go1 in yeast. FEMS Yeast Res 2008; 8:405-13. [DOI: 10.1111/j.1567-1364.2007.00351.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
43
|
Lee JH, Lee BH, Choi SH, Yoon IS, Pyo MK, Shin TJ, Choi WS, Lim Y, Rhim H, Won KH, Lim YW, Choe H, Kim DH, Kim YI, Nah SY. Ginsenoside Rg3 Inhibits Human Kv1.4 Channel Currents by Interacting with the Lys531 Residue. Mol Pharmacol 2007; 73:619-26. [DOI: 10.1124/mol.107.040360] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
44
|
Peters J, Chin CK. Potassium loss is involved in tobacco cell death induced by palmitoleic acid and ceramide. Arch Biochem Biophys 2007; 465:180-6. [PMID: 17662229 DOI: 10.1016/j.abb.2007.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 05/28/2007] [Accepted: 05/30/2007] [Indexed: 12/18/2022]
Abstract
Tobacco cell death induced by palmitoleic acid (16:1), ceramide, and KCN was found to possess features associated with program cell death (PCD), including cell volume decrease, loss of membrane integrity, DNA damage, nuclear and plastid disorganization, and chromatin condensation. Cell volume decrease was found to be caused by loss of intracellular K(+). Ba(2+) was able to prevent the K(+) loss and it also protected the cells from death induced by 16:1 and ceramide but not KCN. The results suggest that K(+) loss is a critical step in plant PCD. The inability of Ba(2+) to prevent cell death was most likely due to its other effects of KCN, i.e., inhibition of cytochrome oxidase in the respiratory chain and generation of reactive oxygen species.
Collapse
Affiliation(s)
- Jeanne Peters
- Department of Plant Biology and Pathology, School of Environmental and Biological, Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | | |
Collapse
|
45
|
Redman PT, He K, Hartnett KA, Jefferson BS, Hu L, Rosenberg PA, Levitan ES, Aizenman E. Apoptotic surge of potassium currents is mediated by p38 phosphorylation of Kv2.1. Proc Natl Acad Sci U S A 2007; 104:3568-73. [PMID: 17360683 PMCID: PMC1805571 DOI: 10.1073/pnas.0610159104] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Kv2.1, the primary delayed rectifying potassium channel in neurons, is extensively regulated by phosphorylation. Previous reports have described Kv2.1 phosphorylation events affecting channel gating and the impact of this process on cellular excitability. Kv2.1, however, also provides the critical exit route for potassium ions during neuronal apoptosis via p38 MAPK-dependent membrane insertion, resulting in a pronounced enhancement of K(+) currents. Here, electrophysiological and viability studies using Kv2.1 channel mutants identify a p38 phosphorylation site at Ser-800 (S800) that is required for Kv2.1 membrane insertion, K(+) current surge, and cell death. In addition, a phospho-specific antibody for S800 detects a p38-dependent increase in Kv2.1 phosphorylation in apoptotic neurons and reveals phosphorylation of S800 in immunopurified channels incubated with active p38. Consequently, phosphorylation of Kv2.1 residue S800 by p38 leads to trafficking and membrane insertion during apoptosis, and remarkably, the absence of S800 phosphorylation is sufficient to prevent completion of the cell death program.
Collapse
Affiliation(s)
| | - Kai He
- Departments of *Neurobiology and
| | | | | | - Linda Hu
- Department of Neurology and Program in Neuroscience, Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Paul A. Rosenberg
- Department of Neurology and Program in Neuroscience, Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Edwin S. Levitan
- Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261; and
| | - Elias Aizenman
- Departments of *Neurobiology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
46
|
Bortner CD, Cidlowski JA. Cell shrinkage and monovalent cation fluxes: role in apoptosis. Arch Biochem Biophys 2007; 462:176-88. [PMID: 17321483 PMCID: PMC1941616 DOI: 10.1016/j.abb.2007.01.020] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Revised: 01/18/2007] [Accepted: 01/23/2007] [Indexed: 12/25/2022]
Abstract
The loss of cell volume or cell shrinkage has been a morphological hallmark of the programmed cell death process known as apoptosis. This isotonic loss of cell volume has recently been term apoptotic volume decrease or AVD to distinguish it from inherent volume regulatory responses that occurs in cells under anisotonic conditions. Recent studies examining the intracellular signaling pathways that result in this unique cellular characteristic have determined that a fundamental movement of ions, particularly monovalent ions, underlie the AVD process and plays an important role on controlling the cell death process. An efflux of intracellular potassium was shown to be a critical aspect of the AVD process, as preventing this ion loss could protect cells from apoptosis. However, potassium plays a complex role as a loss of intracellular potassium has also been shown to be beneficial to the health of the cell. Additionally, the mechanisms that a cell employs to achieve this loss of intracellular potassium vary depending on the cell type and stimulus used to induce apoptosis, suggesting multiple ways exist to accomplish the same goal of AVD. Additionally, sodium and chloride have been shown to play a vital role during cell death in both the signaling and control of AVD in various apoptotic model systems. This review examines the relationship between this morphological change and intracellular monovalent ions during apoptosis.
Collapse
Affiliation(s)
- Carl D Bortner
- The Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Department of Health and Human Services, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
47
|
Grishin A, Li H, Levitan ES, Zaks-Makhina E. Identification of gamma-aminobutyric acid receptor-interacting factor 1 (TRAK2) as a trafficking factor for the K+ channel Kir2.1. J Biol Chem 2006; 281:30104-11. [PMID: 16895905 DOI: 10.1074/jbc.m602439200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To identify proteins that regulate potassium channel activity and expression, we performed functional screening of mammalian cDNA libraries in yeast that express the mammalian K(+) channel Kir2.1. Growth of Kir2.1-expressing yeast in media with low K(+) concentration is a function of K(+) uptake via Kir2.1 channels. Therefore, the host strain was transformed with a human cDNA library, and cDNA clones that rescued growth at low K(+) concentration were selected. One of these clones was identical to the protein of unknown function isolated previously as gamma-aminobutyric acid receptor-interacting factor 1 (GRIF-1) (Beck, M., Brickley, K., Wilkinson, H., Sharma, S., Smith, M., Chazot, P., Pollard, S., and Stephenson, F. (2002) J. Biol. Chem. 277, 30079-30090). GRIF-1 specifically enhanced Kir2.1-dependent growth in yeast and Kir2.1-mediated (86)Rb(+) efflux in HEK293 cells. Quantitative microscopy and flow cytometry analysis of immunolabeled surface Kir2.1 channel showed that GRIF-1 significantly increased the number of Kir2.1 channels in the plasma membrane of COS and HEK293 cells. Physical interaction of Kir2.1 channel and GRIF-1 was demonstrated by co-immunoprecipitation from HEK293 lysates and yeast two-hybrid assay. In vivo association of Kir2.1 and GRIF-1 was demonstrated by co-immunoprecipitation from brain lysate. Yeast two-hybrid assays showed that an N-terminal region of GRIF-1 interacts with a C-terminal region of Kir2.1. These results indicate that GRIF-1 binds to Kir2.1 and facilitates trafficking of this channel to the cell surface.
Collapse
Affiliation(s)
- Anatoly Grishin
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
48
|
Salvador-Recatala V, Kim Y, Zaks-Makhina E, Levitan ES. Voltage-gated k+ channel block by catechol derivatives: defining nonselective and selective pharmacophores. J Pharmacol Exp Ther 2006; 319:758-64. [PMID: 16880198 DOI: 10.1124/jpet.106.107607] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
High-throughput screening led to the identification of a 3-norbornyl derivative of catechol called 48F10 (3-bicyclo[2.2.1]hept-2-yl-benzene-1,2-diol) as a Kv2.1 K(+) channel inhibitor. By virtue of the involvement of Kv2.1 channels in programmed cell death, 48F10 prevents apoptosis in cortical neurons and enterocytes. This uncharged compound acts with an apparent affinity of 1 muM at the tetraethylammonium (TEA) site at the external mouth of the Kv2.1 channel but is ineffective on Kv1.5. Here we investigated the basis of this selectivity with structure-activity studies. We find that catechol (1,2-benzenediol), unlike 48F10, inhibits Kv2.1 currents with a Hill coefficient of 2 and slows channel activation. Furthermore, this inhibition, which requires millimolar concentrations, is unaffected by external TEA or by mutation of the external tyrosine implicated in channel block by TEA and 48F10. In addition, catechol does not distinguish between Kv2.1 and Kv1.5. Thus, catechol acts at conserved sites that are distinct from 48F10. We also tested 11 catechol derivatives based on hydrocarbon adducts including norbornyl substructures, a 48F10 isomer, and a 48F10 diastereomer. These compounds are more potent than catechol, but none replicated the marked selectivity of 48F10 for Kv2.1 over Kv1.5. We conclude that the targeting of 48F10 to the TEA site at the external mouth of the Kv2.1 pore and away from other sites involved in nonselective Kv channel block by catechol requires the norbornyl group in a unique position and orientation on the catechol ring.
Collapse
|
49
|
Friedmann Y, Shriki A, Bennett ER, Golos S, Diskin R, Marbach I, Bengal E, Engelberg D. JX401, A p38α Inhibitor Containing a 4-Benzylpiperidine Motif, Identified via a Novel Screening System in Yeast. Mol Pharmacol 2006; 70:1395-405. [PMID: 16847144 DOI: 10.1124/mol.106.022962] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In vivo screening of compounds for potential pharmacological activity is more advantageous than in vitro screening. In vivo screens eliminate the isolation of compounds that cannot cross biological membranes, are cytotoxic, or are not specific to the target. However, animal-based or even cell-based systems are usually expensive, time-consuming, and laborious. Here we describe the identification of inhibitors of the mitogen-activated protein kinase p38alpha via a high throughput screen using yeast cells. p38alpha is hyperactive in inflammatory diseases, and various indications suggest that its inhibition would reverse inflammation. However, there are currently no p38alpha inhibitors in clinical use. Because the human p38alpha imposes severe growth retardation when expressed in yeast, we screened a library of 40,000 randomly selected small molecules for compounds that would restore a normal growth rate. We identified two compounds; both share a structural motif of 4-benzylpiperidine, and both were shown to be efficient and selective p38alpha inhibitors in vitro. They were also active in mammalian cells, as manifested by their ability to reversibly inhibit myoblast differentiation. Thus, the yeast screen identified efficient and specific p38alpha inhibitors that are capable of crossing biological membranes, are not toxic, and function in mammalian cells. The rapid and cost-efficient high-throughput screening used here could be applied for isolation of inhibitors of various targets.
Collapse
|
50
|
Abstract
Apoptosis in cortical neurons requires efflux of cytoplasmic potassium mediated by a surge in Kv2.1 channel activity. Pharmacological blockade or molecular disruption of these channels in neurons prevents apoptotic cell death, while ectopic expression of Kv2.1 channels promotes apoptosis in non-neuronal cells. Here, we use a cysteine-containing mutant of Kv2.1 and a thiol-reactive covalent inhibitor to demonstrate that the increase in K+ current during apoptosis is due to de novo insertion of functional channels into the plasma membrane. Biotinylation experiments confirmed the delivery of additional Kv2.1 protein to the cell surface following an apoptotic stimulus. Finally, expression of botulinum neurotoxins that cleave syntaxin and synaptosome-associated protein of 25 kDa (SNAP-25) blocked upregulation of surface Kv2.1 channels in cortical neurons, suggesting that target soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins support proapoptotic delivery of K+ channels. These data indicate that trafficking of Kv2.1 channels to the plasma membrane causes the apoptotic surge in K+ current.
Collapse
Affiliation(s)
- SK Pal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15217, USA
| | - K Takimoto
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA
| | - E Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15217, USA
- *Corresponding author: E Aizenman, Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15217, USA., Tel: +412-648-9434; Fax: +412-648-1441; E-mail:
| | - ES Levitan
- Department of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15217, USA
| |
Collapse
|