Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732124

ABSTRACT

Oxytocin, a significant pleiotropic neuropeptide, regulates psychological stress adaptation and social communication, as well as peripheral actions, such as uterine contraction and milk ejection. Recently, a Japanese Kampo medicine called Kamikihito (KKT) has been reported to stimulate oxytocin neurons to induce oxytocin secretion. Two-pore-domain potassium channels (K2P) regulate the resting potential of excitable cells, and their inhibition results in accelerated depolarization that elicits neuronal and endocrine cell activation. We assessed the effects of KKT and 14 of its components on a specific K2P, the potassium channel subfamily K member 2 (TREK-1), which is predominantly expressed in oxytocin neurons in the central nervous system (CNS). KKT inhibited the activity of TREK-1 induced via the channel activator ML335. Six of the 14 components of KKT inhibited TREK-1 activity. Additionally, we identified that 22 of the 41 compounds in the six components exhibited TREK-1 inhibitory effects. In summary, several compounds included in KKT partially activated oxytocin neurons by inhibiting TREK-1. The pharmacological effects of KKT, including antistress effects, may be partially mediated through the oxytocin pathway.


Subject(s)
Neurons , Oxytocin , Potassium Channels, Tandem Pore Domain , Animals , Humans , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Medicine, Kampo , Neurons/metabolism , Neurons/drug effects , Oxytocin/pharmacology , Oxytocin/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors
2.
Nat Commun ; 14(1): 1160, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859433

ABSTRACT

By endowing light control of neuronal activity, optogenetics and photopharmacology are powerful methods notably used to probe the transmission of pain signals. However, costs, animal handling and ethical issues have reduced their dissemination and routine use. Here we report LAKI (Light Activated K+ channel Inhibitor), a specific photoswitchable inhibitor of the pain-related two-pore-domain potassium TREK and TRESK channels. In the dark or ambient light, LAKI is inactive. However, alternating transdermal illumination at 365 nm and 480 nm reversibly blocks and unblocks TREK/TRESK current in nociceptors, enabling rapid control of pain and nociception in intact and freely moving mice and nematode. These results demonstrate, in vivo, the subcellular localization of TREK/TRESK at the nociceptor free nerve endings in which their acute inhibition is sufficient to induce pain, showing LAKI potential as a valuable tool for TREK/TRESK channel studies. More importantly, LAKI gives the ability to reversibly remote-control pain in a non-invasive and physiological manner in naive animals, which has utility in basic and translational pain research but also in in vivo analgesic drug screening and validation, without the need of genetic manipulations or viral infection.


Subject(s)
Pain , Potassium Channels, Tandem Pore Domain , Animals , Mice , Drug Evaluation, Preclinical , Nociceptors , Nematoda , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors
3.
Neuropharmacology ; 224: 109330, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36375694

ABSTRACT

Neuroinflammation, specifically the NLRP3 inflammasome cascade, is a common underlying pathological feature of many neurodegenerative diseases. Evidence suggests that NLRP3 activation involves changes in intracellular K+. Nuclear Enriched Transcript Sort Sequencing (NETSseq), which allows for deep sequencing of purified cell types from human post-mortem brain tissue, demonstrated a highly specific expression of the tandem pore domain halothane-inhibited K+ channel 1 (THIK-1) in microglia compared to other glial and neuronal cell types in the human brain. NETSseq also showed a significant increase of THIK-1 in microglia isolated from cortical regions of brains with Alzheimer's disease (AD) relative to control donors. Herein, we report the discovery and pharmacological characterisation of C101248, the first selective small-molecule inhibitor of THIK-1. C101248 showed a concentration-dependent inhibition of both mouse and human THIK-1 (IC50: ∼50 nM) and was inactive against K2P family members TREK-1 and TWIK-2, and Kv2.1. Whole-cell patch-clamp recordings of microglia from mouse hippocampal slices showed that C101248 potently blocked both tonic and ATP-evoked THIK-1 K+ currents. Notably, C101248 had no effect on other constitutively active resting conductance in slices from THIK-1-depleted mice. In isolated microglia, C101248 prevented NLRP3-dependent release of IL-1ß, an effect not seen in THIK-1-depleted microglia. In conclusion, we demonstrated that inhibiting THIK-1 (a microglia specific gene that is upregulated in brains from donors with AD) using a novel selective modulator attenuates the NLRP3-dependent release of IL-1ß from microglia, which suggests that this channel may be a potential therapeutic target for the modulation of neuroinflammation in AD.


Subject(s)
Alzheimer Disease , Inflammasomes , Potassium Channels, Tandem Pore Domain , Animals , Humans , Mice , Alzheimer Disease/metabolism , Brain/metabolism , Inflammasomes/metabolism , Microglia , Neuroinflammatory Diseases , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors
4.
Cardiovasc Res ; 118(7): 1728-1741, 2022 06 22.
Article in English | MEDLINE | ID: mdl-34028533

ABSTRACT

AIMS: TASK-1 (K2P3.1) two-pore-domain potassium channels are atrial-specific and significantly up-regulated in atrial fibrillation (AF) patients, contributing to AF-related electrical remodelling. Inhibition of TASK-1 in cardiomyocytes of AF patients was shown to counteract AF-related action potential duration shortening. Doxapram was identified as a potent inhibitor of the TASK-1 channel. In this study, we investigated the antiarrhythmic efficacy of doxapram in a porcine model of AF. METHODS AND RESULTS: Doxapram successfully cardioverted pigs with artificially induced episodes of AF. We established a porcine model of persistent AF in domestic pigs via intermittent atrial burst stimulation using implanted pacemakers. All pigs underwent catheter-based electrophysiological investigations prior to and after 14 days of doxapram treatment. Pigs in the treatment group received intravenous administration of doxapram once per day. In doxapram-treated AF pigs, the AF burden was significantly reduced. After 14 days of treatment with doxapram, TASK-1 currents were still similar to values of sinus rhythm animals. Doxapram significantly suppressed AF episodes and normalized cellular electrophysiology by inhibition of the TASK-1 channel. Patch-clamp experiments on human atrial cardiomyocytes, isolated from patients with and without AF could reproduce the TASK-1 inhibitory effect of doxapram. CONCLUSION: Repurposing doxapram might yield a promising new antiarrhythmic drug to treat AF in patients.


Subject(s)
Atrial Fibrillation , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain , Animals , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Doxapram/therapeutic use , Heart Atria/metabolism , Humans , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Swine
5.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Article in English | MEDLINE | ID: mdl-34642249

ABSTRACT

Microglia are the resident immune cells of the central nervous system. They constantly survey the brain parenchyma for redundant synapses, debris, or dying cells, which they remove through phagocytosis. Microglial ramification, motility, and cytokine release are regulated by tonically active THIK-1 K+ channels on the microglial plasma membrane. Here, we examined whether these channels also play a role in phagocytosis. Using pharmacological blockers and THIK-1 knockout (KO) mice, we found that a lack of THIK-1 activity approximately halved both microglial phagocytosis and marker levels for the lysosomes that degrade phagocytically removed material. These changes may reflect a decrease of intracellular [Ca2+]i activity, which was observed when THIK-1 activity was reduced, since buffering [Ca2+]i reduced phagocytosis. Less phagocytosis is expected to result in impaired pruning of synapses. In the hippocampus, mice lacking THIK-1 expression had an increased number of anatomically and electrophysiologically defined glutamatergic synapses during development. This resulted from an increased number of presynaptic terminals, caused by impaired removal by THIK-1 KO microglia. The dependence of synapse number on THIK-1 K+ channels, which control microglial surveillance and phagocytic ability, implies that changes in the THIK-1 expression level in disease states may contribute to altering neural circuit function.


Subject(s)
Microglia/metabolism , Potassium Channels, Tandem Pore Domain/physiology , Synapses/physiology , Animals , Calcium/metabolism , Female , Male , Mice , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Rats , Rats, Sprague-Dawley , Synapses/metabolism
6.
Behav Brain Res ; 413: 113446, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34224765

ABSTRACT

TREK-1 channels are expressed in small nociceptive dorsal root ganglion (DRG) neurons where they participate in acute inflammatory and neuropathic pain. However, the role of TREK-1 in persistent pain is not well understood. The aim of this study was to investigate the local peripheral and spinal participation of TREK-1 in formalin-induced acute and long-lasting nociceptive hypersensitivity. Local peripheral or intrathecal pre-treatment with spadin, selective blocker of TREK-1, increased acute flinching behavior and secondary mechanical allodynia and hyperalgesia behavior observed 6 days after formalin injection. Local peripheral or intrathecal pre-treatment with BL-1249, selective opener of TREK-1, decreased long-lasting secondary mechanical allodynia and hyperalgesia induced by formalin. Pre-treatment with BL-1249 prevented the pro-nociceptive effect of spadin on acute nociception and long-lasting mechanical allodynia and hyperalgesia in rats. Pre-treatment with two recombinant channels that produce a high TREK-1 current, S300A and S333A (non-phosphorylated state of TREK-1), reduced formalin-induced acute pain and long-lasting mechanical allodynia and hyperalgesia. Besides, post-treatment with S300A, S333A or BL-1249 reversed long-lasting mechanical allodynia and hyperalgesia induced by formalin. Formalin increased TREK-1 expression at 1 and 6 days in DRG and dorsal spinal cord in rats, whereas that it increased c-fos expression at the DRG. Intrathecal repeated transfection of rats with S300A and S333A or injection with BL-1249 reduced formalin-induced enhanced c-fos expression. Data suggest that TREK-1 activity at peripheral and spinal sites reduces neuronal excitability in the process of acute and long-lasting nociception induced by formalin in rats.


Subject(s)
Disinfectants/pharmacology , Formaldehyde/pharmacology , Ganglia, Spinal , Hyperalgesia , Nociceptive Pain , Potassium Channels, Tandem Pore Domain/metabolism , Spinal Cord , Animals , Disease Models, Animal , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Nociceptive Pain/chemically induced , Nociceptive Pain/drug therapy , Nociceptive Pain/metabolism , Peptides/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/drug effects , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , Tetrahydronaphthalenes/pharmacology , Tetrazoles/pharmacology
7.
Molecules ; 26(13)2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34202296

ABSTRACT

TASK channels belong to the two-pore-domain potassium (K2P) channels subfamily. These channels modulate cellular excitability, input resistance, and response to synaptic stimulation. TASK-channel inhibition led to membrane depolarization. TASK-3 is expressed in different cancer cell types and neurons. Thus, the discovery of novel TASK-3 inhibitors makes these bioactive compounds very appealing to explore new cancer and neurological therapies. TASK-3 channel blockers are very limited to date, and only a few heterofused compounds have been reported in the literature. In this article, we combined a pharmacophore hypothesis with molecular docking to address for the first time the rational design, synthesis, and evaluation of 5-(indol-2-yl)pyrazolo[3,4-b]pyridines as a novel family of human TASK-3 channel blockers. Representative compounds of the synthesized library were assessed against TASK-3 using Fluorometric imaging plate reader-Membrane Potential assay (FMP). Inhibitory properties were validated using two-electrode voltage-clamp (TEVC) methods. We identified one active hit compound (MM-3b) with our systematic pipeline, exhibiting an IC50 ≈ 30 µM. Molecular docking models suggest that compound MM-3b binds to TASK-3 at the bottom of the selectivity filter in the central cavity, similar to other described TASK-3 blockers such as A1899 and PK-THPP. Our in silico and experimental studies provide a new tool to predict and design novel TASK-3 channel blockers.


Subject(s)
Molecular Docking Simulation , Potassium Channel Blockers , Potassium Channels, Tandem Pore Domain , Pyridines , Humans , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry
8.
Am J Physiol Cell Physiol ; 321(1): C158-C175, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34038243

ABSTRACT

In whole cell patch clamp recordings, it was discovered that normal human adrenal zona glomerulosa (AZG) cells express members of the three major families of K+ channels. Among these are a two-pore (K2P) leak-type and a G protein-coupled, inwardly rectifying (GIRK) channel, both inhibited by peptide hormones that stimulate aldosterone secretion. The K2P current displayed properties identifying it as TREK-1 (KCNK2). This outwardly rectifying current was activated by arachidonic acid and inhibited by angiotensin II (ANG II), adrenocorticotrophic hormone (ACTH), and forskolin. The activation and inhibition of TREK-1 was coupled to AZG cell hyperpolarization and depolarization, respectively. A second K2P channel, TASK-1 (KCNK3), was expressed at a lower density in AZG cells. Human AZG cells also express inwardly rectifying K+ current(s) (KIR) that include quasi-instantaneous and time-dependent components. This is the first report demonstrating the presence of KIR in whole cell recordings from AZG cells of any species. The time-dependent current was selectively inhibited by ANG II, and ACTH, identifying it as a G protein-coupled (GIRK) channel, most likely KIR3.4 (KCNJ5). The quasi-instantaneous KIR current was not inhibited by ANG II or ACTH and may be a separate non-GIRK current. Finally, AZG cells express a voltage-gated, rapidly inactivating K+ current whose properties identified as KV1.4 (KCNA4), a conclusion confirmed by Northern blot. These findings demonstrate that human AZG cells express K2P and GIRK channels whose inhibition by ANG II and ACTH is likely coupled to depolarization-dependent secretion. They further demonstrate that human AZG K+ channels differ fundamentally from the widely adopted rodent models for human aldosterone secretion.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Angiotensin II/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Kv1.4 Potassium Channel/genetics , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Zona Glomerulosa/metabolism , Adolescent , Adult , Aldosterone/biosynthesis , Arachidonic Acid/pharmacology , Autopsy , Child , Colforsin/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression , Humans , Kv1.4 Potassium Channel/antagonists & inhibitors , Kv1.4 Potassium Channel/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Middle Aged , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/metabolism , Primary Cell Culture , Zona Glomerulosa/cytology , Zona Glomerulosa/drug effects
9.
SLAS Discov ; 26(3): 460-469, 2021 03.
Article in English | MEDLINE | ID: mdl-33334229

ABSTRACT

Voltage-gated ion channels produce rapid transmembrane currents responsible for action potential generation and propagation at the neuronal, muscular, and cardiac levels. They represent attractive clinical targets because their altered firing frequency is often the hallmark of pathological signaling leading to several neuromuscular disorders. Therefore, a method to study their functioning upon repeated triggers at different frequencies is desired to develop new drug molecules selectively targeting pathological phenotype. Optogenetics provides powerful tools for millisecond switch of cellular excitability in contactless, physiological, and low-cost settings. Nevertheless, its application to large-scale drug-screening operations is still limited by long processing time (due to sequential well read), rigid flashing pattern, lack of online compound addition, or high consumable costs of existing methods. Here, we developed a method that enables simultaneous analysis of 384-well plates with optical pacing, fluorescence recording, and liquid injection. We used our method to deliver programmable millisecond-switched depolarization through light-activated opsin in concomitance with continuous optical recording by a fluorescent indicator. We obtained 384-well pacing of recombinant voltage-activated sodium or calcium channels, as well as induced pluripotent stem cell (iPSC)-derived cardiomyocytes, in all-optical parallel settings. Furthermore, we demonstrated the use-dependent behavior of known ion channel blockers by optogenetic pacing at normal or pathological firing frequencies, obtaining very good signal reproducibility and accordance with electrophysiology data. Our method provides a novel physiological approach to study frequency-dependent drug behavior using reversible programmable triggers. The all-optical parallel settings combined with contained operational costs make our method particularly suited for large-scale drug-screening campaigns as well as cardiac liability studies.


Subject(s)
Biological Assay , Calcium Channel Blockers/pharmacology , Optogenetics/methods , Potassium Channel Blockers/pharmacology , Algal Proteins/antagonists & inhibitors , Algal Proteins/genetics , Algal Proteins/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Line , Chlamydomonas reinhardtii , Fluorescent Dyes/chemistry , Gene Expression , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ion Channel Gating/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Optical Imaging/methods , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Rhodopsin/antagonists & inhibitors , Rhodopsin/genetics , Rhodopsin/metabolism
10.
J Neurochem ; 157(6): 2039-2054, 2021 06.
Article in English | MEDLINE | ID: mdl-33006141

ABSTRACT

PKC and PKA phosphorylation inhibit TREK-1 channels downstream of Gs protein-coupled receptor activation in vitro. However, the role of phosphorylation of TREK-1 in neuropathic pain is unknown. The purpose of this study was to investigate whether altered TREK-1 channel function by PKA and PKC modulators contributes to antiallodynia in neuropathic rats. Furthermore, we investigated if the in vitro described sites for PKC and PKA phosphorylation (S300 and S333, respectively) participate in the modulation of TREK-1 in naïve and neuropathic rats. L5/L6 spinal nerve ligation (SNL) induced tactile allodynia. Intrathecal injection of BL-1249 (TREK-1 activator) reversed nerve injury-induced tactile allodynia, whereas spadin (TREK-1 blocker) produced tactile allodynia in naïve rats and reversed the antiallodynic effect induced by BL-1249 in neuropathic rats. Intrathecal administration of rottlerin or Rp-cAMPs (PKC and PKA inhibitors, respectively) enhanced the antiallodynia observed with BL-1249 in neuropathic rats. In contrast, pretreatment with PdBu or forskolin (PKC and PKA activators, respectively) reduced the BL-1249-induced antiallodynia. Intrathecal injection of two high-activity TREK-1 recombinant channels, using a in vivo transfection method with lipofectamine, with mutations at PKC/PKA phosphosites (S300A and S333A) reversed tactile allodynia in neuropathic rats, with no effect in naïve rats. In contrast, transfection of two low-activity TREK-1 recombinant channels with phosphomimetic mutations at those sites (S300D and S333D) produced tactile allodynia in naïve rats and interfered with antiallodynic effects of rottlerin/BL-1249 or Rp-cAMPs/BL-1249. Data suggest that TREK-1 channel activity can be dynamically tuned in vivo by PKC/PKA to provoke allodynia and modulate its antiallodynic role in neuropathic pain.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Neuralgia/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Protein Kinase C/metabolism , Animals , Female , Injections, Spinal , Mice , Neuralgia/drug therapy , Pain Measurement/methods , Peptides/administration & dosage , Phosphorylation/drug effects , Phosphorylation/physiology , Potassium Channels, Tandem Pore Domain/agonists , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Rats , Rats, Wistar , Tetrahydronaphthalenes/administration & dosage , Tetrazoles/administration & dosage
11.
CNS Neurosci Ther ; 27(2): 220-232, 2021 02.
Article in English | MEDLINE | ID: mdl-32864894

ABSTRACT

INTRODUCTION: The two-pore domain potassium channel TREK-1 is a member of background K+ channels that are thought to provide baseline regulation of membrane excitability. Recent studies have highlighted the putative role of TREK-1 in the action of antidepressants, and its antagonists might be potentially effective antidepressants. However, the mechanisms underlying the actions of TREK-1 are not yet fully understood. METHODS: The expression of TREK-1 was examined in a mouse model of chronic unpredictable mild stress (CUMS) using immunoblotting. Neuron-specific genetic manipulation of TREK-1 was performed through adeno-associated virus. Behavioral tests were performed to evaluate depression-related behaviors. Electrophysiological recordings were used to evaluate synaptic plasticity. Golgi staining was used to examine neuroplasticity. RESULTS: TREK-1 expression was increased in the mouse hippocampus after CUMS. Knockdown of TREK-1 in hippocampal neurons significantly attenuated depressive-like behaviors and prevented the decrease of CUMS-induced synaptic proteins in mice. Further examination indicated that neuron-specific knockdown of TREK-1 in the hippocampus prevented stress-induced impairment of glutamatergic synaptic transmission in the CA1 region. Moreover, chronic TREK-1 inhibition protected against CUMS-induced depressive-like behaviors and impairment of synaptogenesis in the hippocampus. CONCLUSION: Our results indicate a role for TREK-1 in the modulation of synaptic plasticity in a mouse model of depression. These findings will provide insight into the pathological mechanism of depression and further evidence for a novel target for antidepressant treatment.


Subject(s)
Depression/genetics , Hippocampus/physiology , Neuronal Plasticity/physiology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Animals , Bridged Bicyclo Compounds/administration & dosage , Depression/drug therapy , Depression/psychology , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Oxazoles/administration & dosage , Peptides/administration & dosage , RNA, Small Interfering/administration & dosage
12.
Int J Mol Sci ; 21(24)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348878

ABSTRACT

Astrocytes, the most abundant cell type in the brain, are non-excitable cells and play critical roles in brain function. Mature astrocytes typically exhibit a linear current-voltage relationship termed passive conductance, which is believed to enable astrocytes to maintain potassium homeostasis in the brain. We previously demonstrated that TWIK-1/TREK-1 heterodimeric channels mainly contribute to astrocytic passive conductance. However, the molecular identity of astrocytic passive conductance is still controversial and needs to be elucidated. Here, we report that spadin, an inhibitor of TREK-1, can dramatically reduce astrocytic passive conductance in brain slices. A series of gene silencing experiments demonstrated that spadin-sensitive currents are mediated by TWIK-1/TREK-1 heterodimeric channels in cultured astrocytes and hippocampal astrocytes from brain slices. Our study clearly showed that TWIK-1/TREK-1-heterodimeric channels can act as the main molecular machinery of astrocytic passive conductance, and suggested that spadin can be used as a specific inhibitor to control astrocytic passive conductance.


Subject(s)
Astrocytes/physiology , Brain/physiology , Gene Expression Regulation/drug effects , Peptides/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Protein Multimerization , Animals , Astrocytes/cytology , Astrocytes/drug effects , Brain/cytology , Brain/drug effects , Female , Male , Mice , Mice, Inbred C57BL
13.
J Med Chem ; 63(19): 10972-10983, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32877186

ABSTRACT

Modulators can be designed to stabilize the inactive and active states of ion channels, but whether intermediate (IM) states of channel gating are druggable remains underexplored. In this study, using molecular dynamics simulations of the TWIK-related potassium channel 1 (TREK-1) channel, a two-pore domain potassium channel, we captured an IM state during the transition from the down (inactive) state to the up (active-like) state. The IM state contained a druggable allosteric pocket that was not present in the down or up state. Drug design targeting the pocket led to the identification of the TKIM compound as an inhibitor of TREK-1. Using integrated methods, we verified that TKIM binds to the pocket of the IM state of TREK-1, which differs from the binding of common inhibitors, which bind to channels in the inactive state. Overall, this study identified an allosteric ligand-binding site and a new mechanistic inhibitor for TREK-1, suggesting that IM states of ion channels may be promising druggable targets for use in discovering allosteric modulators.


Subject(s)
Drug Design , Ion Channel Gating/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Allosteric Site , Animals , CHO Cells , Cricetulus , Humans , Ligands , Molecular Dynamics Simulation , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/metabolism
14.
Mol Pharmacol ; 98(2): 143-155, 2020 08.
Article in English | MEDLINE | ID: mdl-32616523

ABSTRACT

The two-pore domain potassium channel (K2P-channel) THIK-1 has several predicted protein kinase A (PKA) phosphorylation sites. In trying to elucidate whether THIK-1 is regulated via PKA, we expressed THIK-1 channels in a mammalian cell line (CHO cells) and used the phosphodiesterase inhibitor 3-isobutyl-1-methyl-xanthine (IBMX) as a pharmacological tool to induce activation of PKA. Using the whole-cell patch-clamp recording, we found that THIK-1 currents were inhibited by application of IBMX with an IC50 of 120 µM. Surprisingly, intracellular application of IBMX or of the second messenger cAMP via the patch pipette had no effect on THIK-1 currents. In contrast, extracellular application of IBMX produced a rapid and reversible inhibition of THIK-1. In patch-clamp experiments with outside-out patches, THIK-1 currents were also inhibited by extracellular application of IBMX. Expression of THIK-1 channels in Xenopus oocytes was used to compare wild-type channels with mutated channels. Mutation of the putative PKA phosphorylation sites did not change the inhibitory effect of IBMX on THIK-1 currents. Mutational analysis of all residues of the (extracellular) helical cap of THIK-1 showed that mutation of the arginine residue at position 92, which is in the linker between cap helix 2 and pore helix 1, markedly reduced the inhibitory effect of IBMX. This flexible linker region, which is unique for each K2P-channel subtype, may be a possible target of channel-specific blockers. SIGNIFICANCE STATEMENT: The potassium channel THIK-1 is strongly expressed in the central nervous system. We studied the effect of 3-isobutyl-1-methyl-xanthine (IBMX) on THIK-1 currents. IBMX inhibits breakdown of cAMP and thus activates protein kinase A (PKA). Surprisingly, THIK-1 current was inhibited when IBMX was applied from the extracellular side of the membrane, but not from the intracellular side. Our results suggest that IBMX binds directly to the channel and that the inhibition of THIK-1 current was not related to activation of PKA.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Arginine/genetics , Binding Sites/drug effects , CHO Cells , Cricetulus , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mutation , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Rats , Xenopus
15.
Mol Biol (Mosk) ; 54(3): 457-468, 2020.
Article in Russian | MEDLINE | ID: mdl-32492009

ABSTRACT

TWIK-related acid-sensitive potassium channel-1 (TASK-1) is a "leak" potassium channel sensitive to extracellular protons. It contributes to setting the resting potential in mammalian neurons. TASK-1 channels are widely expressed in respiratory-related neurons in the central nervous system. Inhibition of TASK-1 by extracellular acidosis can depolarize and increase the excitability of these cells. Here we describe the distribution of TASK-1 in the rat brainstem and show that TASK-1 mRNAs are present in respiratory-related nuclei in the ventrolateral medulla, which have been proposed as neural substrates for central chemo-reception in rats. After inhalation of 8% CO2 for 30 and 60 min, TASK-1 mRNA levels in positive-expression neurons were remarkably upregulated. Injection of the TASK-1 blocker anandamide (AEA) into the rat lateral cerebral ventricle, showed a significant excitement of respiratory at 10 min posttreatment, with a marked decrease in inspiratory and expiratory durations and an increased frequency of respiration. We suggest that TASK-1 channel may serve as a chemosensor for in central respiration and may contribute to pH-sensitive respiratory effects. TASK-1 channel might be an attractive candidate for sensing H^(+)/CO2 in several respiratory-related nuclei in the brainstem. It is likely that TASK-1 participates in pH-sensitive chemical regulation in the respiratory center under physiological and pathological conditions.


Subject(s)
Nerve Tissue Proteins/physiology , Neurons/physiology , Potassium Channels, Tandem Pore Domain/physiology , Respiratory Center/physiology , Animals , Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , Hydrogen-Ion Concentration , Membrane Potentials , Nerve Tissue Proteins/antagonists & inhibitors , Polyunsaturated Alkamides/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Rats
16.
Nature ; 582(7812): 443-447, 2020 06.
Article in English | MEDLINE | ID: mdl-32499642

ABSTRACT

TWIK-related acid-sensitive potassium (TASK) channels-members of the two pore domain potassium (K2P) channel family-are found in neurons1, cardiomyocytes2-4 and vascular smooth muscle cells5, where they are involved in the regulation of heart rate6, pulmonary artery tone5,7, sleep/wake cycles8 and responses to volatile anaesthetics8-11. K2P channels regulate the resting membrane potential, providing background K+ currents controlled by numerous physiological stimuli12-15. Unlike other K2P channels, TASK channels are able to bind inhibitors with high affinity, exceptional selectivity and very slow compound washout rates. As such, these channels are attractive drug targets, and TASK-1 inhibitors are currently in clinical trials for obstructive sleep apnoea and atrial fibrillation16. In general, potassium channels have an intramembrane vestibule with a selectivity filter situated above and a gate with four parallel helices located below; however, the K2P channels studied so far all lack a lower gate. Here we present the X-ray crystal structure of TASK-1, and show that it contains a lower gate-which we designate as an 'X-gate'-created by interaction of the two crossed C-terminal M4 transmembrane helices at the vestibule entrance. This structure is formed by six residues (243VLRFMT248) that are essential for responses to volatile anaesthetics10, neurotransmitters13 and G-protein-coupled receptors13. Mutations within the X-gate and the surrounding regions markedly affect both the channel-open probability and the activation of the channel by anaesthetics. Structures of TASK-1 bound to two high-affinity inhibitors show that both compounds bind below the selectivity filter and are trapped in the vestibule by the X-gate, which explains their exceptionally low washout rates. The presence of the X-gate in TASK channels explains many aspects of their physiological and pharmacological behaviour, which will be beneficial for the future development and optimization of TASK modulators for the treatment of heart, lung and sleep disorders.


Subject(s)
Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/chemistry , Anesthetics/pharmacology , Animals , Crystallography, X-Ray , Electric Conductivity , Female , Humans , Ion Channel Gating/drug effects , Models, Molecular , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Xenopus laevis
17.
Biomed Pharmacother ; 129: 110383, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32563149

ABSTRACT

Withaferin A (WFA), a C5,C6-epoxy steroidal lactone isolated from the medicinal plant Withania somnifera (L.) Dunal, inhibits growth of tumor cells in different cancer types. However, the mechanisms underlying the effect of WFA on tumor cells are not fully understood. In the present study, we evaluated the blockade of TASK-3 channels by WFA in TASK-3-expressing HEK-293 cells. Explore if the WFA-mediated TASK-3 blockade can be used as a pharmacological tool to decrease the cell viability in cancer cells. A combination of functional experiments (patch-clamp, gene downregulation, overexpression and pharmacological inhibition) and molecular docking analysis were used to get insights into the mechanism by which the inhibition of TASK-3 by WFA affects the growth and viability of cancer cells. Withaferin A was found to inhibit the activity of TASK-3 channels. The inhibitory effect of Withaferin A on TASK-3 potassium currents was dose-dependent and independent of voltage. Molecular modeling studies identified putative WFA-binding sites in TASK-3 channel involved the channel blockade. In agreements with the molecular modeling predictions, mutation of residues F125 to A (F125A), L197 to V (L197 V) and the double mutant F125A-L197 V markedly decreased the WFA-induced inhibition of TASK-3. Finally, the cytotoxic effect of WFA was tested in MDA-MB-231 human breast cancer cells transfected with TASK-3 or shRNA that decreases TASK-3 expression. Together, our results show that the cytotoxic effect of WFA on fully transformed MDA-MB-231 cells depends on the expression of TASK-3. Herein, we also provide insights into the mechanism of TASK-3 inhibition by WFA.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Withanolides/pharmacology , Antineoplastic Agents, Phytogenic/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Membrane Potentials , Potassium Channel Blockers/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Protein Binding , Signal Transduction , Withanolides/metabolism
18.
J Am Heart Assoc ; 9(10): e015751, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32390491

ABSTRACT

Background The tandem of P domains in a weak inward rectifying K+ channel (TWIK)-related acid-sensitive K+ channel (TASK-1; hK2P3.1) two-pore-domain potassium channel was recently shown to regulate the atrial action potential duration. In the human heart, TASK-1 channels are specifically expressed in the atria. Furthermore, upregulation of atrial TASK-1 currents was described in patients suffering from atrial fibrillation (AF). We therefore hypothesized that TASK-1 channels represent an ideal target for antiarrhythmic therapy of AF. In the present study, we tested the antiarrhythmic effects of the high-affinity TASK-1 inhibitor A293 on cardioversion in a porcine model of paroxysmal AF. Methods and Results Heterologously expressed human and porcine TASK-1 channels are blocked by A293 to a similar extent. Patch clamp measurements from isolated human and porcine atrial cardiomyocytes showed comparable TASK-1 currents. Computational modeling was used to investigate the conditions under which A293 would be antiarrhythmic. German landrace pigs underwent electrophysiological studies under general anesthesia. Paroxysmal AF was induced by right atrial burst stimulation. After induction of AF episodes, intravenous administration of A293 restored sinus rhythm within cardioversion times of 177±63 seconds. Intravenous administration of A293 resulted in significant prolongation of the atrial effective refractory period, measured at cycle lengths of 300, 400 and 500 ms, whereas the surface ECG parameters and the ventricular effective refractory period lengths remained unchanged. Conclusions Pharmacological inhibition of atrial TASK-1 currents exerts antiarrhythmic effects in vivo as well as in silico, resulting in acute cardioversion of paroxysmal AF. Taken together, these experiments indicate the therapeutic potential of A293 for AF treatment.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Sulfonamides/pharmacology , ortho-Aminobenzoates/pharmacology , Animals , Atrial Fibrillation/diagnosis , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Disease Models, Animal , Electrocardiography , Electrophysiologic Techniques, Cardiac , Female , Humans , Male , Membrane Potentials/drug effects , Molecular Docking Simulation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Proof of Concept Study , Refractory Period, Electrophysiological/drug effects , Sus scrofa , Time Factors , Xenopus laevis
19.
Cell Chem Biol ; 27(5): 511-524.e4, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32059793

ABSTRACT

The trinuclear ruthenium amine ruthenium red (RuR) inhibits diverse ion channels, including K2P potassium channels, TRPs, the calcium uniporter, CALHMs, ryanodine receptors, and Piezos. Despite this extraordinary array, there is limited information for how RuR engages targets. Here, using X-ray crystallographic and electrophysiological studies of an RuR-sensitive K2P, K2P2.1 (TREK-1) I110D, we show that RuR acts by binding an acidic residue pair comprising the "Keystone inhibitor site" under the K2P CAP domain archway above the channel pore. We further establish that Ru360, a dinuclear ruthenium amine not known to affect K2Ps, inhibits RuR-sensitive K2Ps using the same mechanism. Structural knowledge enabled a generalizable design strategy for creating K2P RuR "super-responders" having nanomolar sensitivity. Together, the data define a "finger in the dam" inhibition mechanism acting at a novel K2P inhibitor binding site. These findings highlight the polysite nature of K2P pharmacology and provide a new framework for K2P inhibitor development.


Subject(s)
Coloring Agents/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Ruthenium Compounds/pharmacology , Ruthenium Red/pharmacology , Amines/chemistry , Amines/pharmacology , Animals , Coloring Agents/chemistry , Crystallography, X-Ray , Mice , Molecular Docking Simulation , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Ruthenium/chemistry , Ruthenium/pharmacology , Ruthenium Compounds/chemistry , Ruthenium Red/chemistry
20.
Circ Arrhythm Electrophysiol ; 12(9): e007465, 2019 09.
Article in English | MEDLINE | ID: mdl-31514528

ABSTRACT

BACKGROUND: Despite an increasing understanding of atrial fibrillation (AF) pathophysiology, translation into mechanism-based treatment options is lacking. In atrial cardiomyocytes of patients with chronic AF, expression, and function of tandem of P domains in a weak inward rectifying TASK-1 (K+ channel-related acid-sensitive K+ channel-1) (K2P3.1) atrial-specific 2-pore domain potassium channels is enhanced, resulting in action potential duration shortening. TASK-1 channel inhibition prevents action potential duration shortening to maintain values observed among sinus rhythm subjects. The present preclinical study used a porcine AF model to evaluate the antiarrhythmic efficacy of TASK-1 inhibition by adeno-associated viral anti-TASK-1-siRNA (small interfering RNA) gene transfer. METHODS: AF was induced in domestic pigs by atrial burst stimulation via implanted pacemakers. Adeno-associated viral vectors carrying anti-TASK-1-siRNA were injected into both atria to suppress TASK-1 channel expression. After the 14-day follow-up period, porcine cardiomyocytes were isolated from right and left atrium, followed by electrophysiological and molecular characterization. RESULTS: AF was associated with increased TASK-1 transcript, protein and ion current levels leading to shortened action potential duration in atrial cardiomyocytes compared to sinus rhythm controls, similar to previous findings in humans. Anti-TASK-1 adeno-associated viral application significantly reduced AF burden in comparison to untreated AF pigs. Antiarrhythmic effects of anti-TASK-1-siRNA were associated with reduction of TASK-1 currents and prolongation of action potential durations in atrial cardiomyocytes to sinus rhythm values. Conclusions Adeno-associated viral-based anti-TASK-1 gene therapy suppressed AF and corrected cellular electrophysiological remodeling in a porcine model of AF. Suppression of AF through selective reduction of TASK-1 currents represents a new option for antiarrhythmic therapy.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/genetics , Atrial Remodeling/physiology , Gene Expression Regulation , Genetic Therapy/methods , Heart Atria/physiopathology , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Action Potentials/physiology , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/therapy , Disease Models, Animal , Electrocardiography , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/biosynthesis , RNA/genetics , Rats , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...