1
|
Li XT. The involvement of K + channels in depression and pharmacological effects of antidepressants on these channels. Transl Psychiatry 2024; 14:411. [PMID: 39358318 PMCID: PMC11447029 DOI: 10.1038/s41398-024-03069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
Depression is a common and complex psychiatric illness with multiple clinical symptoms, even leading to the disability and suicide. Owing to the partial understanding of the pathogenesis of depressive-like disorders, available pharmacotherapeutic strategies are developed mainly based on the "monoamine hypothesis", resulting in a limited effectiveness and a number of adverse effects in the clinical practice. The concept of multiple pathogenic factors be helpful for clarifying the etiology of depression and developing the antidepressants. It is well documented that K+ channels serve crucial roles in modulating the neuronal excitability and neurotransmitter release in the brain, and abnormality of these channels participated in the pathogenic process of diverse central nervous system (CNS) pathologies, such as seizure and Alzheimer's disease (AD). The clinical and preclinical evidence also delineates that the involvement of several types of K+ channels in depressive-like behaviors appear to be evident, suggesting these channels being one of the multiple factors in the etiology of this debilitating disorder. Emerging data manifest that diverse antidepressants impact distinct K+ channels, such as Kv, Kir and K2P, meaning the functioning of these drug via a "multi-target" manner. On the other hand, the scenario of antidepressants impinging K+ channels could render an alternative interpretation for the pharmacological effectiveness and numerous side effects in clinical trials. Furthermore, these channels serve to be considered as a "druggable target" to develop novel therapeutic compound to antagonize this psychiatry.
Collapse
Affiliation(s)
- Xian-Tao Li
- School of Medicine, Jingchu University of Technology, Jingmen, China.
- Research group of Neurological and Metabolic Disease, School of Medicine, Jingchu University of Technology, Jingmen, China.
| |
Collapse
|
2
|
Park SJ, Silic MR, Staab PL, Chen J, Zackschewski EL, Zhang G. Evolution of two-pore domain potassium channels and their gene expression in zebrafish embryos. Dev Dyn 2024; 253:722-749. [PMID: 38270285 PMCID: PMC11269526 DOI: 10.1002/dvdy.690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/10/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND The two-pore domain potassium (K2P) channels are a major type of potassium channels that maintain the cell membrane potential by conducting passive potassium leak currents independent of voltage change. They play prominent roles in multiple physiological processes, including neuromodulation, perception of pain, breathing and mood control, and response to volatile anesthetics. Mutations in K2P channels have been linked to many human diseases, such as neuronal and cardiovascular disorders and cancers. Significant progress has been made to understand their protein structures, physiological functions, and pharmacological modifiers. However, their expression and function during embryonic development remain largely unknown. RESULTS We employed the zebrafish model and identified 23 k2p genes using BLAST search and gene cloning. We first analyzed vertebrate K2P channel evolution by phylogenetic and syntenic analyses. Our data revealed that the six subtypes of the K2P genes have already evolved in invertebrates long before the emergence of vertebrates. Moreover, the vertebrate K2P gene number increased, most likely due to two whole-genome duplications. Furthermore, we examined zebrafish k2p gene expression during early embryogenesis by in situ hybridization. Each subgroup's genes showed similar but distinct gene expression domains with some exceptions. Most of them were expressed in neural tissues consistent with their known function of neural excitability regulation. However, a few k2p genes were expressed temporarily in specific tissues or organs, suggesting that these K2P channels may be needed for embryonic development. CONCLUSIONS Our phylogenetic and developmental analyses of K2P channels shed light on their evolutionary history and potential roles during embryogenesis related to their physiological functions and human channelopathies.
Collapse
Affiliation(s)
- Sung Jun Park
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
| | - Martin R. Silic
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
| | - Peyton L. Staab
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
| | - Jiapei Chen
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
| | - Ethan L. Zackschewski
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
| | - GuangJun Zhang
- Department of Comparative Pathobiology, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
- Purdue University Center for Cancer Research, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
- Purdue Institute for Inflammation, Immunology and Infectious Diseases (PI4D), Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
- Purdue Institute for Integrative Neuroscience, Purdue University, 625 Harrison Street, West Lafayette, Indiana. 47906. USA
| |
Collapse
|
3
|
Delanne-Cuménal M, Lamoine S, Meleine M, Aissouni Y, Prival L, Fereyrolles M, Barbier J, Cercy C, Boudieu L, Schopp J, Lazdunski M, Eschalier A, Lolignier S, Busserolles J. The TREK-1 potassium channel is involved in both the analgesic and anti-proliferative effects of riluzole in bone cancer pain. Biomed Pharmacother 2024; 176:116887. [PMID: 38852511 DOI: 10.1016/j.biopha.2024.116887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND The metastasis of tumors into bone tissue typically leads to intractable pain that is both very disabling and particularly difficult to manage. We investigated here whether riluzole could have beneficial effects for the treatment of prostate cancer-induced bone pain and how it could influence the development of bone metastasis. METHODS We used a bone pain model induced by intratibial injection of human PC3 prostate cancer cells into male SCID mice treated or not with riluzole administered in drinking water. We also used riluzole in vitro to assess its possible effect on PC3 cell viability and functionality, using patch-clamp. RESULTS Riluzole had a significant preventive effect on both evoked and spontaneous pain involving the TREK-1 potassium channel. Riluzole did not interfere with PC3-induced bone loss or bone remodeling in vivo. It also significantly decreased PC3 cell viability in vitro. The antiproliferative effect of riluzole is correlated with a TREK-1-dependent membrane hyperpolarization in these cells. CONCLUSION The present data suggest that riluzole could be very useful to manage evoked and spontaneous hypersensitivity in cancer-induced bone pain and has no significant adverse effect on cancer progression.
Collapse
Affiliation(s)
- Mélissa Delanne-Cuménal
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Sylvain Lamoine
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Mathieu Meleine
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Youssef Aissouni
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Laetitia Prival
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Mathilde Fereyrolles
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Julie Barbier
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Christine Cercy
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Ludivine Boudieu
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Julien Schopp
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Michel Lazdunski
- Université de Nice Sophia Antipolis, Valbonne 06560, France; CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, 660 Route des Lucioles Sophia Antipolis, Valbonne 06560, France
| | - Alain Eschalier
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France; Institut Analgesia, Faculté de Médecine, BP38, Clermont-Ferrand 63001, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France
| | - Jérôme Busserolles
- Université Clermont Auvergne, Inserm, CHU Clermont-Ferrand, Neuro-Dol, Clermont-Ferrand F63000, France.
| |
Collapse
|
4
|
Rajendran R, Krishnan R, Kim JO, Oh MJ. Regulatory effects of potassium channel blockers on potassium channel genes upon nervous necrosis virus infection in sevenband grouper Hyporthodus septumfasciatus. Gene 2024; 890:147815. [PMID: 37739197 DOI: 10.1016/j.gene.2023.147815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Ion channels in fishes regulate the flow of important ions that play an active role in the excitation and transmission of impulses through neuronal cells. Specific housekeeping genes translates into proteins and selectively permeabilize and facilitate ion crossover transmissions. Potassium (K+) channels play a crucial role in a wide range of functions such as cell volume regulation, hormone secretion, synaptic transmission and muscle contraction. The dysfunction of ion channels result in channelopathies, which hinder critical cellular activities. Recent studies have indicated that viral pathogens tend to regulate cellular ion channels for entry into host cells. Hence, the present study aimed to elucidate the role of K+ channels during nervous necrosis virus (NNV) infections in the sevenband grouper (Hyporthodus septumfasciatus). Real-time PCR with the standardized potassium genes revealed the downregulation of potassium two pore domain channel subfamily member - KCNK10, KCNK9, KCNK2, and KCNK1 genes post infection at both 17 °C and 25 °C whereas an upregulation was noted in the case of gill tissues. SMART analysis revealed a transmembrane region in all genes. Multiple sequence alignment using MultAlin and phylogenetic analysis revealed true homology of potassium genes with other higher vertebrates. In vitro and in vivo challenge study of NNV using Tetra ethyl ammonium (TEA) as potential drug showed inverse relation to that of viral replication and a corresponding downregulation of K+ channel gene expression was observed which was further confirmed by an immunofluorescence assay. These findings indicate that K+ channels play a crucial role during viral infection. Moreover, the observed downregulation can be related to rapid endocytosis resulting from recycling endosomes during a viral infection. Hence, further studies are warranted to better understand the role of K+ channel genes during NNV infection.
Collapse
Affiliation(s)
- Rahul Rajendran
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 50626, Republic of Korea
| | - Rahul Krishnan
- Department of Aquatic Animal Health Management, Faculty of Fisheries, Kerala University of Fisheries and Ocean Studies, Ernakulam, Kerala, India
| | - Jong-Oh Kim
- Department of Microbiology, Pukyong National University, Busan, Republic of Korea
| | - Myung-Joo Oh
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 50626, Republic of Korea.
| |
Collapse
|
5
|
Genta PR, Taranto-Montemurro L. Task Accomplished: Promising Effects of a New Topical Potassium Channel Antagonist in OSA. Chest 2023; 163:749-750. [PMID: 37031981 DOI: 10.1016/j.chest.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 04/11/2023] Open
Affiliation(s)
- Pedro R Genta
- Heart Institute, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
6
|
Lenzini L, Caroccia B, Seccia TM, Rossi GP. Peptidergic G Protein-Coupled Receptor Regulation of Adrenal Function: Bench to Bedside and Back. Endocr Rev 2022; 43:1038-1050. [PMID: 35436330 DOI: 10.1210/endrev/bnac011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 11/19/2022]
Abstract
An altered secretion of adrenocortical and adrenomedullary hormones plays a role in the clinical syndromes of primary aldosteronism (PA), Cushing, and pheochromocytoma. Moreover, an altered production of adrenocortical hormones and/or an abnormal release of factors by the adrenal medulla are involved in several other diseases, including high blood pressure, congestive heart failure, liver cirrhosis, nephrotic syndrome, primary reninism, renovascular hypertension, Addison disease, Bartter, Gitelman, and virilization syndromes. Understanding the regulation of adrenal function and the interactions between adrenal cortex and medulla is, therefore, the prerequisite for mechanistic understanding of these disorders. Accumulating evidence indicates that the modulation of adrenal hormone biosynthesis is a process far more complex than originally thought, as it involves several factors, each cooperating with the other. Moreover, the tight vascular and neural interconnections between the adrenal cortex and medulla underlie physiologically relevant autocrine/paracrine interactions involving several peptides. Besides playing a pathophysiological role in common adrenal diseases, these complex mechanisms could intervene also in rare diseases, such as pheochromocytoma concomitant with adrenal Cushing or with PA, and PA co-occurring with Cushing, through mechanisms that remain to be fully understood at the molecular levels. Heterodimerization of G protein-coupled receptors (GPCRs) induced by peptide signaling is a further emerging new modulatory mechanism capable of finely tuning adrenal hormones synthesis and release. In this review we will examine current knowledge on the role of peptides that act via GPCRs in the regulation of adrenal hormone secretion with a particular focus on autocrine-paracrine signals.
Collapse
Affiliation(s)
- Livia Lenzini
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| | - Brasilina Caroccia
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| | - Teresa Maria Seccia
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| | - Gian Paolo Rossi
- Emergency Medicine Unit, Center for blood pressure disorders -Regione Veneto and Specialized Center of Excellence for Hypertension of the European Society of Hypertension, Department of Medicine-DIMED, University of Padua, 35126 Padua, Italy
| |
Collapse
|
7
|
Li XT. The modulation of potassium channels by estrogens facilitates neuroprotection. Front Cell Dev Biol 2022; 10:998009. [PMID: 36393851 PMCID: PMC9643774 DOI: 10.3389/fcell.2022.998009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/11/2022] [Indexed: 08/31/2023] Open
Abstract
Estrogens, the sex hormones, have the potential to govern multiple cellular functions, such as proliferation, apoptosis, differentiation, and homeostasis, and to exert numerous beneficial influences for the cardiovascular system, nervous system, and bones in genomic and/or non-genomic ways. Converging evidence indicates that estrogens serve a crucial role in counteracting neurodegeneration and ischemic injury; they are thereby being considered as a potent neuroprotectant for preventing neurological diseases such as Alzheimer's disease and stroke. The underlying mechanism of neuroprotective effects conferred by estrogens is thought to be complex and multifactorial, and it remains obscure. It is well established that the K+ channels broadly expressed in a variety of neural subtypes determine the essential physiological features of neuronal excitability, and dysfunction of these channels is closely associated with diverse brain deficits, such as ataxia and epilepsy. A growing body of evidence supports a neuroprotective role of K+ channels in malfunctions of nervous tissues, with the channels even being a therapeutic target in clinical trials. As multitarget steroid hormones, estrogens also regulate the activity of distinct K+ channels to generate varying biological actions, and accumulated data delineate that some aspects of estrogen-mediated neuroprotection may arise from the impact on multiple K+ channels, including Kv, BK, KATP, and K2P channels. The response of these K+ channels after acute or chronic exposure to estrogens may oppose pathological abnormality in nervous cells, which serves to extend our understanding of these phenomena.
Collapse
Affiliation(s)
- Xian-Tao Li
- School of Medicine, Guizhou University, Guiyang, China
- Department of Neuroscience, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
8
|
Glial-derived neurotrophic factor regulates the expression of TREK2 in rat primary sensory neurons leading to attenuation of axotomy-induced neuropathic pain. Exp Neurol 2022; 357:114190. [PMID: 35907583 DOI: 10.1016/j.expneurol.2022.114190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/12/2022] [Accepted: 07/24/2022] [Indexed: 11/24/2022]
Abstract
TREK2 is a member of the 2-pore domain family of K+ channels (K2P) preferentially expressed by unmyelinated, slow-conducting and non-peptidergic isolectin B4-binding (IB4+) primary sensory neurons of the dorsal root ganglia (DRG). IB4+ neurons depend on the glial-derived neurotrophic factor (GDNF) family of ligands (GFL's) to maintain their phenotype. In our previous work, we demonstrated that 7 days after spinal nerve axotomy (SNA) of the L5 DRG, TREK2 moves away from the cell membrane resulting in a more depolarised resting membrane potential (Em). Given that axotomy deprives DRG neurons from peripherally-derived GFL's, we hypothesized that they might control the expression of TREK2. Using a combination of immunohistochemistry, immunocytochemistry, western blotting, in vivo pharmacological manipulation and behavioral tests we examined the ability of the GFL's (GDNF, neurturin and artemin) and their selective receptors (GFRα1, GFRα2 and GFRα3) to regulate the expression and function of TREK2 in the DRG. We found that TREK2 correlated strongly with the three receptors normally and ipsilaterally for all GFR's after SNA. GDNF, but not NGF, neurturin or artemin up-regulated the expression of TREK2 in cultured DRG neurons. In vivo continuous, subcutaneous administration of GDNF restored the subcellular distribution of TREK2 ipsilaterally and reversed mechanical and cold allodynia 7 days after SNA. This is the first demonstration that GDNF controls the expression of a K2P channel in nociceptors. As TREK2 controls the Em of C-nociceptors affecting their excitability, our finding has therapeutic potential in the treatment of chronic pain.
Collapse
|
9
|
Liu S, Guo P, Wang K, Zhang S, Li Y, Shen J, Mei L, Ye Y, Zhang Q, Yang H. General Pharmacological Activation Mechanism of K + Channels Bypassing Channel Gates. J Med Chem 2022; 65:10285-10299. [PMID: 35878013 DOI: 10.1021/acs.jmedchem.1c02115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Under the known pharmacological activation mechanisms, activators allosterically or directly open potassium channel gates. However, herein, molecular dynamics simulations on TREK-1, a member of the channel class gated at the filter, suggested that negatively charged activators act with a gate-independent mechanism where compounds increase currents by promoting ions passing through the central cavity. Then, based on studies of KCNQ2, we uncovered that this noncanonical activation mechanism is shared by the other channel class gated at the helix-bundle crossing. Rational drug design found a novel KCNQ2 agonist, CLE030, which stably binds to the central cavity. Functional analysis, molecular dynamics simulations, and calculations of the potential of mean force revealed that the carbonyl oxygen of CLE030 influences permeant ions in the central cavity to contribute to its activation effects. Together, this study discovered a ligand-to-ion activation mechanism for channels that bypasses their gates and thus is conserved across subfamilies with different gates.
Collapse
Affiliation(s)
- Shijie Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peipei Guo
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Kun Wang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shaoying Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ya Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Juwen Shen
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lianghe Mei
- Suzhou Institute of Drug Innovation, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, China
| | - Yangliang Ye
- Suzhou AlphaMa Biotechnology Co., Ltd., Suzhou, Jiangsu 215123, China
| | - Qiansen Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
10
|
Li XT. Beneficial effects of carvedilol modulating potassium channels on the control of glucose. Biomed Pharmacother 2022; 150:113057. [PMID: 35658228 DOI: 10.1016/j.biopha.2022.113057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/25/2022] Open
Abstract
The increased prevalence of hypertensive patients with type 2 diabetes mellitus (T2DM) is evident worldwide, leading to a higher risk of cardiovascular disease onset, which is substantially associated with disabilities and mortality in the clinic. In order to achieve the satisfyingly clinical outcomes and prognosis, the comprehensive therapies have been conducted with a beneficial effect on both blood pressure and glucose homeostasis, and clinical trials reveal that some kind of antihypertensive drugs such as angiotensin converting enzyme inhibitors (ACE-I) may, at least in part, meet the dual requirement during the disease management. As a nonselective β-blocker, carvedilol is employed for treating many cardiovascular diseases in clinical practice, including hypertension, angina pectoris and heart failure, and also exhibit the effectiveness for glycemic control and insulin resistance. Apart from alleviating sympathetic nervous system activity, several causes, such as lowering oxygen reactive species, may contribute to the effects of carvedilol on controlling plasma glucose levels, suggesting a feature of this drug having multiple targets. Interestingly, numerous distinct K+ channels expressed in pancreatic β-cells and peripheral insulin-sensitive tissues, which play a sentential role in glucose metabolism, are subjected to extensive modulation of carvdilol, establishing a linkage between K+ channels and drug's effects on the control of glucose. A variety of evidence shows that the impact of carvedilol on different K+ channels, including Kv, KAch, KATP and K2 P, can lead to positive influences for glucose homeostasis, contributing to its clinical beneficial effectiveness in treatment of hypertensive patients with T2DM. This review focus on the control of plasma glucose conferred by carvedilol modulation on K+ channels, providing the novel mechanistic explanation for drug's actions.
Collapse
Affiliation(s)
- Xian-Tao Li
- Department of Neuroscience, South-Central University for Nationalities, Wuhan 430074, China; School of Medicine, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
11
|
García-Morales V, Gento-Caro Á, Portillo F, Montero F, González-Forero D, Moreno-López B. Lysophosphatidic Acid and Several Neurotransmitters Converge on Rho-Kinase 2 Signaling to Manage Motoneuron Excitability. Front Mol Neurosci 2021; 14:788039. [PMID: 34938160 PMCID: PMC8685439 DOI: 10.3389/fnmol.2021.788039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/05/2021] [Indexed: 01/18/2023] Open
Abstract
Intrinsic membrane excitability (IME) sets up neuronal responsiveness to synaptic drive. Several neurotransmitters and neuromodulators, acting through G-protein-coupled receptors (GPCRs), fine-tune motoneuron (MN) IME by modulating background K+ channels TASK1. However, intracellular partners linking GPCRs to TASK1 modulation are not yet well-known. We hypothesized that isoform 2 of rho-kinase (ROCK2), acting as downstream GPCRs, mediates adjustment of MN IME via TASK1. Electrophysiological recordings were performed in hypoglossal MNs (HMNs) obtained from adult and neonatal rats, neonatal knockout mice for TASK1 (task1–/–) and TASK3 (task3–/–, the another highly expressed TASK subunit in MNs), and primary cultures of embryonic spinal cord MNs (SMNs). Small-interfering RNA (siRNA) technology was also used to knockdown either ROCK1 or ROCK2. Furthermore, ROCK activity assays were performed to evaluate the ability of various physiological GPCR ligands to stimulate ROCK. Microiontophoretically applied H1152, a ROCK inhibitor, and siRNA-induced ROCK2 knockdown both depressed AMPAergic, inspiratory-related discharge activity of adult HMNs in vivo, which mainly express the ROCK2 isoform. In brainstem slices, intracellular constitutively active ROCK2 (aROCK2) led to H1152-sensitive HMN hyper-excitability. The aROCK2 inhibited pH-sensitive and TASK1-mediated currents in SMNs. Conclusively, aROCK2 increased IME in task3–/–, but not in task1–/– HMNs. MN IME was also augmented by the physiological neuromodulator lysophosphatidic acid (LPA) through a mechanism entailing Gαi/o-protein stimulation, ROCK2, but not ROCK1, activity and TASK1 inhibition. Finally, two neurotransmitters, TRH, and 5-HT, which are both known to increase MN IME by TASK1 inhibition, stimulated ROCK2, and depressed background resting currents via Gαq/ROCK2 signaling. These outcomes suggest that LPA and several neurotransmitters impact MN IME via Gαi/o/Gαq-protein-coupled receptors, downstream ROCK2 activation, and subsequent inhibition of TASK1 channels.
Collapse
Affiliation(s)
- Victoria García-Morales
- GRUpo de NEuroDEgeneración y NeurorREparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| | - Ángela Gento-Caro
- GRUpo de NEuroDEgeneración y NeurorREparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| | - Federico Portillo
- GRUpo de NEuroDEgeneración y NeurorREparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| | - Fernando Montero
- GRUpo de NEuroDEgeneración y NeurorREparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - David González-Forero
- GRUpo de NEuroDEgeneración y NeurorREparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| | - Bernardo Moreno-López
- GRUpo de NEuroDEgeneración y NeurorREparación (GRUNEDERE), Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Cádiz, Spain
| |
Collapse
|
12
|
Huang W, Ke Y, Zhu J, Liu S, Cong J, Ye H, Guo Y, Wang K, Zhang Z, Meng W, Gao TM, Luhmann HJ, Kilb W, Chen R. TRESK channel contributes to depolarization-induced shunting inhibition and modulates epileptic seizures. Cell Rep 2021; 36:109404. [PMID: 34289346 DOI: 10.1016/j.celrep.2021.109404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/19/2021] [Accepted: 06/23/2021] [Indexed: 11/18/2022] Open
Abstract
Glutamatergic and GABAergic synaptic transmission controls excitation and inhibition of postsynaptic neurons, whereas activity of ion channels modulates neuronal intrinsic excitability. However, it is unclear how excessive neuronal excitation affects intrinsic inhibition to regain homeostatic stability under physiological or pathophysiological conditions. Here, we report that a seizure-like sustained depolarization can induce short-term inhibition of hippocampal CA3 neurons via a mechanism of membrane shunting. This depolarization-induced shunting inhibition (DShI) mediates a non-synaptic, but neuronal intrinsic, short-term plasticity that is able to suppress action potential generation and postsynaptic responses by activated ionotropic receptors. We demonstrate that the TRESK channel significantly contributes to DShI. Disruption of DShI by genetic knockout of TRESK exacerbates the sensitivity and severity of epileptic seizures of mice, whereas overexpression of TRESK attenuates seizures. In summary, these results uncover a type of homeostatic intrinsic plasticity and its underlying mechanism. TRESK might represent a therapeutic target for antiepileptic drugs.
Collapse
Affiliation(s)
- Weiyuan Huang
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yue Ke
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianping Zhu
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuai Liu
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jin Cong
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hailin Ye
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanwu Guo
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Kewan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenhai Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Center for Precision Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tian-Ming Gao
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Collaborative Innovation Center for Brain Science, Southern Medical University, Guangzhou 510515, China
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz 55120, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz 55120, Germany.
| | - Rongqing Chen
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
13
|
Mitochondrial Function and Anesthetic Sensitivity in the Mouse Spinal Cord. Anesthesiology 2021; 134:901-914. [PMID: 33909880 DOI: 10.1097/aln.0000000000003794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Ndufs4 knockout (KO) mice are defective in mitochondrial complex I function and hypersensitive to inhibition of spinal cord-mediated response to noxious stimuli by volatile anesthetics. It was hypothesized that, compared to wild-type, synaptic or intrinsic neuronal function is hypersensitive to isoflurane in spinal cord slices from knockout mice. METHODS Neurons from slices of the vestibular nucleus, central medial thalamus, and spinal cord from wild-type and the global Ndufs4 knockout were patch clamped. Unstimulated synaptic and intrinsic neuronal characteristics were measured in response to isoflurane. Norfluoxetine was used to block TREK channel conductance. Cholinergic cells were labeled with tdTomato. RESULTS All values are reported as means and 95% CIs. Spontaneous synaptic activities were not different between the mutant and control. Isoflurane (0.6%; 0.25 mM; Ndufs4[KO] EC95) increased the holding current in knockout (ΔHolding current, 126 pA [95% CI, 99 to 152 pA]; ΔHolding current P < 0.001; n = 21) but not wild-type (ΔHolding current, 2 7 pA [95% CI, 9 to 47 pA]; ΔHolding current, P = 0.030; n = 25) spinal cord slices. Knockout and wild-type ΔHolding currents were significantly different (P < 0.001). Changes comparable to those in the knockout were seen in the wild type only in 1.8% (0.74 mM) isoflurane (ΔHolding current, 72 pA [95% CI, 43 to 97 pA]; ΔHolding current, P < 0.001; n = 13), the control EC95. Blockade of action potentials indicated that the increased holding current in the knockout was not dependent on synaptic input (ΔHolding current, 154 pA [95% CI, 99 to 232 pA]; ΔHolding current, P = 0.506 compared to knockout without blockade; n = 6). Noncholinergic neurons mediated the increase in holding current sensitivity in Ndufs4 knockout. The increased currents were blocked by norfluoxetine. CONCLUSIONS Isoflurane increased an outwardly rectifying potassium current in ventral horn neurons of the Ndufs4(KO) mouse at a concentration much lower than in controls. Noncholinergic neurons in the spinal cord ventral horn mediated the effect. Presynaptic functions in Ndufs4(KO) slices were not hypersensitive to isoflurane. These data link anesthetic sensitivity, mitochondrial function, and postsynaptic channel activity. EDITOR’S PERSPECTIVE
Collapse
|
14
|
Rossi GP, Lenzini L, Caroccia B, Rossitto G, Seccia TM. Angiotensin peptides in the regulation of adrenal cortical function. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The adrenal cortex plays a key role in the regulation of metabolism, salt and water homeostasis and sex differentiation by synthesizing glucocorticoid, mineralocorticoid and androgen hormones. Evidence exists that angiotensin II regulates adrenocortical function and it has been contended that angiotensin peptides of the non-canonical branch of the renin angiotensin system (RAS) might also modulate steroidogenesis in adrenals. Thus, the aim of this review is to examine the role of the RAS, and particularly of the angiotensin peptides and their receptors, in the regulation of adrenocortical hormones with particular focus on aldosterone production.
Collapse
Affiliation(s)
- Gian Paolo Rossi
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Livia Lenzini
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Brasilina Caroccia
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Giacomo Rossitto
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| | - Teresa Maria Seccia
- Emergency Medicine Unit and European Society of Hypertension Specialized Center of Excellence for Hypertension, Department of Medicine-DIMED, University of Padua, Italy
| |
Collapse
|
15
|
Brouns I, Verckist L, Pintelon I, Timmermans JP, Adriaensen D. Pulmonary Sensory Receptors. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2021; 233:1-65. [PMID: 33950466 DOI: 10.1007/978-3-030-65817-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Inge Brouns
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium.
| | - Line Verckist
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| | - Dirk Adriaensen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| |
Collapse
|
16
|
Bortolozzi A, Manashirov S, Chen A, Artigas F. Oligonucleotides as therapeutic tools for brain disorders: Focus on major depressive disorder and Parkinson's disease. Pharmacol Ther 2021; 227:107873. [PMID: 33915178 DOI: 10.1016/j.pharmthera.2021.107873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/05/2021] [Indexed: 12/25/2022]
Abstract
Remarkable advances in understanding the role of RNA in health and disease have expanded considerably in the last decade. RNA is becoming an increasingly important target for therapeutic intervention; therefore, it is critical to develop strategies for therapeutic modulation of RNA function. Oligonucleotides, including antisense oligonucleotide (ASO), small interfering RNA (siRNA), microRNA mimic (miRNA), and anti-microRNA (antagomir) are perhaps the most direct therapeutic strategies for addressing RNA. Among other mechanisms, most oligonucleotide designs involve the formation of a hybrid with RNA that promotes its degradation by activation of endogenous enzymes such as RNase-H (e.g., ASO) or the RISC complex (e.g. RNA interference - RNAi for siRNA and miRNA). However, the use of oligonucleotides for the treatment of brain disorders is seriously compromised by two main limitations: i) how to deliver oligonucleotides to the brain compartment, avoiding the action of peripheral RNAses? and once there, ii) how to target specific neuronal populations? We review the main molecular pathways in major depressive disorder (MDD) and Parkinson's disease (PD), and discuss the challenges associated with the development of novel oligonucleotide therapeutics. We pay special attention to the use of conjugated ligand-oligonucleotide approach in which the oligonucleotide sequence is covalently bound to monoamine transporter inhibitors (e.g. sertraline, reboxetine, indatraline). This strategy allows their selective accumulation in the monoamine neurons of mice and monkeys after their intranasal or intracerebroventricular administration, evoking preclinical changes predictive of a clinical therapeutic action after knocking-down disease-related genes. In addition, recent advances in oligonucleotide therapeutic clinical trials are also reviewed.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain; Institut d'Investigacions August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain.
| | - Sharon Manashirov
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain; miCure Therapeutics LTD., Tel-Aviv, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Francesc Artigas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain; Institut d'Investigacions August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain
| |
Collapse
|
17
|
Natale AM, Deal PE, Minor DL. Structural Insights into the Mechanisms and Pharmacology of K 2P Potassium Channels. J Mol Biol 2021; 433:166995. [PMID: 33887333 PMCID: PMC8436263 DOI: 10.1016/j.jmb.2021.166995] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 01/10/2023]
Abstract
Leak currents, defined as voltage and time independent flows of ions across cell membranes, are central to cellular electrical excitability control. The K2P (KCNK) potassium channel class comprises an ion channel family that produces potassium leak currents that oppose excitation and stabilize the resting membrane potential in cells in the brain, cardiovascular system, immune system, and sensory organs. Due to their widespread tissue distribution, K2Ps contribute to many physiological and pathophysiological processes including anesthesia, pain, arrythmias, ischemia, hypertension, migraine, intraocular pressure regulation, and lung injury responses. Structural studies of six homomeric K2Ps have established the basic architecture of this channel family, revealed key moving parts involved in K2P function, uncovered the importance of asymmetric pinching and dilation motions in the K2P selectivity filter (SF) C-type gate, and defined two K2P structural classes based on the absence or presence of an intracellular gate. Further, a series of structures characterizing K2P:modulator interactions have revealed a striking polysite pharmacology housed within a relatively modestly sized (~70 kDa) channel. Binding sites for small molecules or lipids that control channel function are found at every layer of the channel structure, starting from its extracellular side through the portion that interacts with the membrane bilayer inner leaflet. This framework provides the basis for understanding how gating cues sensed by different channel parts control function and how small molecules and lipids modulate K2P activity. Such knowledge should catalyze development of new K2P modulators to probe function and treat a wide range of disorders.
Collapse
Affiliation(s)
- Andrew M Natale
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Parker E Deal
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience University of California, San Francisco, CA 94158, USA; Molecular Biophysics and Integrated Bio-imaging Division Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
18
|
Lee H, Lolicato M, Arrigoni C, Minor DL. Production of K 2P2.1 (TREK-1) for structural studies. Methods Enzymol 2021; 653:151-188. [PMID: 34099170 DOI: 10.1016/bs.mie.2021.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
K2P (KCNK) potassium channels form 'background' or 'leak' currents that are important for controlling cell excitability in the brain, cardiovascular system, and somatosensory neurons. K2P2.1 (TREK-1) is one of the founding members of this family and one of the first well-characterized polymodal ion channels capable of responding to a variety of physical and chemical gating cues. Of the six K2P subfamilies, the thermo-and mechano-sensitive TREK subfamily comprising K2P2.1 (TREK-1), K2P4.1 (TRAAK), and K2P10.1 (TREK-2) is the first to have structures determined for each subfamily member. These structural studies have revealed key architectural features that provide a framework for understanding how gating cues sensed by different channel elements converge on the K2P selectivity filter C-type gate. TREK family structural studies have also revealed numerous sites where small molecules or lipids bind and affect channel function. This rich structural landscape provides the framework for probing K2P function and for the development of new K2P-directed agents. Such molecules may be useful for affecting processes where TREK channels are important such as anesthesia, pain, arrythmia, ischemia, migraine, intraocular pressure, and lung injury. Production of high quality protein samples is key to addressing new questions about K2P function and pharmacology. Here, we present methods for producing pure K2P2.1 (TREK-1) suitable for advancing towards these goals through structural and biochemical studies.
Collapse
Affiliation(s)
- Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Cristina Arrigoni
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA, United States; California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, United States; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, United States; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
19
|
Kraft M, Büscher A, Wiedmann F, L’hoste Y, Haefeli WE, Frey N, Katus HA, Schmidt C. Current Drug Treatment Strategies for Atrial Fibrillation and TASK-1 Inhibition as an Emerging Novel Therapy Option. Front Pharmacol 2021; 12:638445. [PMID: 33897427 PMCID: PMC8058608 DOI: 10.3389/fphar.2021.638445] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia with a prevalence of up to 4% and an upwards trend due to demographic changes. It is associated with an increase in mortality and stroke incidences. While stroke risk can be significantly reduced through anticoagulant therapy, adequate treatment of other AF related symptoms remains an unmet medical need in many cases. Two main treatment strategies are available: rate control that modulates ventricular heart rate and prevents tachymyopathy as well as rhythm control that aims to restore and sustain sinus rhythm. Rate control can be achieved through drugs or ablation of the atrioventricular node, rendering the patient pacemaker-dependent. For rhythm control electrical cardioversion and pharmacological cardioversion can be used. While electrical cardioversion requires fasting and sedation of the patient, antiarrhythmic drugs have other limitations. Most antiarrhythmic drugs carry a risk for pro-arrhythmic effects and are contraindicated in patients with structural heart diseases. Furthermore, catheter ablation of pulmonary veins can be performed with its risk of intraprocedural complications and varying success. In recent years TASK-1 has been introduced as a new target for AF therapy. Upregulation of TASK-1 in AF patients contributes to prolongation of the action potential duration. In a porcine model of AF, TASK-1 inhibition by gene therapy or pharmacological compounds induced cardioversion to sinus rhythm. The DOxapram Conversion TO Sinus rhythm (DOCTOS)-Trial will reveal whether doxapram, a potent TASK-1 inhibitor, can be used for acute cardioversion of persistent and paroxysmal AF in patients, potentially leading to a new treatment option for AF.
Collapse
Affiliation(s)
- Manuel Kraft
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Antonius Büscher
- Clinic for Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Felix Wiedmann
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Yannick L’hoste
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Hugo A. Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Bachmann M, Rossa A, Antoniazzi G, Biasutto L, Carrer A, Campagnaro M, Leanza L, Gonczi M, Csernoch L, Paradisi C, Mattarei A, Zoratti M, Szabo I. Synthesis and cellular effects of a mitochondria-targeted inhibitor of the two-pore potassium channel TASK-3. Pharmacol Res 2021; 164:105326. [PMID: 33338625 DOI: 10.1016/j.phrs.2020.105326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/03/2020] [Accepted: 11/23/2020] [Indexed: 01/25/2023]
Abstract
The two-pore potassium channel TASK-3 has been shown to localize to both the plasma membrane and the mitochondrial inner membrane. TASK-3 is highly expressed in melanoma and breast cancer cells and has been proposed to promote tumor formation. Here we investigated whether pharmacological modulation of TASK-3, and specifically of mitochondrial TASK-3 (mitoTASK-3), had any effect on cancer cell survival and mitochondrial physiology. A novel, mitochondriotropic version of the specific TASK-3 inhibitor IN-THPP has been synthesized by addition of a positively charged triphenylphosphonium moiety. While IN-THPP was unable to induce apoptosis, mitoIN-THPP decreased survival of breast cancer cells and efficiently killed melanoma lines, which we show to express mitoTASK-3. Cell death was accompanied by mitochondrial membrane depolarization and fragmentation of the mitochondrial network, suggesting a role of the channel in the maintenance of the correct function of this organelle. In accordance, cells treated with mitoIN-THPP became rapidly depleted of mitochondrial ATP which resulted in activation of the AMP-dependent kinase AMPK. Importantly, cell survival was not affected in mouse embryonic fibroblasts and the effect of mitoIN-THPP was less pronounced in human melanoma cells stably knocked down for TASK-3 expression, indicating a certain degree of selectivity of the drug both for pathological cells and for the channel. In addition, mitoIN-THPP inhibited cancer cell migration to a higher extent than IN-THPP in two melanoma cell lines. In summary, our results point to the importance of mitoTASK-3 for melanoma cell survival and migration.
Collapse
Affiliation(s)
| | - Andrea Rossa
- Department of Chemical Sciences, University of Padua, Italy
| | | | - Lucia Biasutto
- CNR Institute of Neuroscience, Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy
| | - Andrea Carrer
- Department of Biology, University of Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy
| | | | - Luigi Leanza
- Department of Biology, University of Padua, Italy
| | - Monika Gonczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Hungary
| | - Laszlo Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Hungary
| | | | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Mario Zoratti
- CNR Institute of Neuroscience, Padua, Italy; Department of Biomedical Sciences, University of Padua, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padua, Italy; CNR Institute of Neuroscience, Padua, Italy.
| |
Collapse
|
21
|
Deardorff AS, Romer SH, Fyffe RE. Location, location, location: the organization and roles of potassium channels in mammalian motoneurons. J Physiol 2021; 599:1391-1420. [DOI: 10.1113/jp278675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/08/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Adam S. Deardorff
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
- Department of Neurology and Internal Medicine, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| | - Shannon H. Romer
- Odyssey Systems Environmental Health Effects Laboratory, Navy Medical Research Unit‐Dayton Wright‐Patterson Air Force Base OH 45433 USA
| | - Robert E.W. Fyffe
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| |
Collapse
|
22
|
Wiedmann F, Beyersdorf C, Zhou XB, Kraft M, Foerster KI, El-Battrawy I, Lang S, Borggrefe M, Haefeli WE, Frey N, Schmidt C. The Experimental TASK-1 Potassium Channel Inhibitor A293 Can Be Employed for Rhythm Control of Persistent Atrial Fibrillation in a Translational Large Animal Model. Front Physiol 2021; 11:629421. [PMID: 33551849 PMCID: PMC7858671 DOI: 10.3389/fphys.2020.629421] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/30/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Upregulation of the two-pore-domain potassium channel TASK-1 (hK2 P 3.1) was recently described in patients suffering from atrial fibrillation (AF) and resulted in shortening of the atrial action potential. In the human heart, TASK-1 channels facilitate repolarization and are specifically expressed in the atria. In the present study, we tested the antiarrhythmic effects of the experimental ion channel inhibitor A293 that is highly affine for TASK-1 in a porcine large animal model of persistent AF. METHODS Persistent AF was induced in German landrace pigs by right atrial burst stimulation via implanted pacemakers using a biofeedback algorithm over 14 days. Electrophysiological and echocardiographic investigations were performed before and after the pharmacological treatment period. A293 was intravenously administered once per day. After a treatment period of 14 days, atrial cardiomyocytes were isolated for patch clamp measurements of currents and atrial action potentials. Hemodynamic consequences of TASK-1 inhibition were measured upon acute A293 treatment. RESULTS In animals with persistent AF, the A293 treatment significantly reduced the AF burden (6.5% vs. 95%; P < 0.001). Intracardiac electrophysiological investigations showed that the atrial effective refractory period was prolonged in A293 treated study animals, whereas, the QRS width, QT interval, and ventricular effective refractory periods remained unchanged. A293 treatment reduced the upregulation of the TASK-1 current as well as the shortening of the action potential duration caused by AF. No central nervous side effects were observed. A mild but significant increase in pulmonary artery pressure was observed upon acute TASK-1 inhibition. CONCLUSION Pharmacological inhibition of atrial TASK-1 currents exerts in vivo antiarrhythmic effects that can be employed for rhythm control in a porcine model of persistent AF. Care has to be taken as TASK-1 inhibition may increase pulmonary artery pressure levels.
Collapse
Affiliation(s)
- Felix Wiedmann
- Department of Cardiology, Heidelberg University, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, Heidelberg University, Heidelberg, Germany
| | - Christoph Beyersdorf
- Department of Cardiology, Heidelberg University, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, Heidelberg University, Heidelberg, Germany
| | - Xiao-Bo Zhou
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- First Department of Medicine, University Medical Center, Mannheim University, Mannheim, Germany
| | - Manuel Kraft
- Department of Cardiology, Heidelberg University, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, Heidelberg University, Heidelberg, Germany
| | - Kathrin I. Foerster
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University, Heidelberg, Germany
| | - Ibrahim El-Battrawy
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- First Department of Medicine, University Medical Center, Mannheim University, Mannheim, Germany
| | - Siegfried Lang
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- First Department of Medicine, University Medical Center, Mannheim University, Mannheim, Germany
| | - Martin Borggrefe
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- First Department of Medicine, University Medical Center, Mannheim University, Mannheim, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Heidelberg University, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, Heidelberg University, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, Heidelberg University, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
23
|
Abstract
Two-pore domain potassium channels are formed by subunits that each contain two pore-loops moieties. Whether the channels are expressed in yeast or the human central nervous system, two subunits come together to form a single potassium selective pore. TOK1, the first two-domain channel was cloned from Saccharomyces cerevisiae in 1995 and soon thereafter, 15 distinct K2P subunits were identified in the human genome. The human K2P channels are stratified into six K2P subfamilies based on sequence as well as physiological or pharmacological similarities. Functional K2P channels pass background (or "leak") K+ currents that shape the membrane potential and excitability of cells in a broad range of tissues. In the years since they were first described, classical functional assays, latterly coupled with state-of-the-art structural and computational studies have revealed the mechanistic basis of K2P channel gating in response to specific physicochemical or pharmacological stimuli. The growing appreciation that K2P channels can play a pivotal role in the pathophysiology of a growing spectrum of diseases makes a compelling case for K2P channels as targets for drug discovery. Here, we summarize recent advances in unraveling the structure, function, and pharmacology of the K2P channels.
Collapse
Affiliation(s)
- Jordie M Kamuene
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Yu Xu
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Leigh D Plant
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
24
|
Functional Exploration of the Pulmonary NEB ME. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2021; 233:31-67. [PMID: 33950469 DOI: 10.1007/978-3-030-65817-5_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
25
|
Herbrechter R, Beltrán LR, Ziemba PM, Titt S, Lashuk K, Gottemeyer A, Levermann J, Hoffmann KM, Beltrán M, Hatt H, Störtkuhl KF, Werner M, Gisselmann G. Effect of 158 herbal remedies on human TRPV1 and the two-pore domain potassium channels KCNK2, 3 and 9. J Tradit Complement Med 2020; 10:446-453. [PMID: 32953560 PMCID: PMC7484967 DOI: 10.1016/j.jtcme.2020.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 11/22/2022] Open
Abstract
Background and aim Herbal medicines are used to treat a broad number of maladies. However, the pharmacological profile of most remedies is poorly understood. We investigated the effect of herbal remedies from kampo, traditional Chinese medicine (TCM) and other phytotherapies on human two-pore domain potassium channels (KCNK channels; TREK-1, TASK-1 and TASK-3) as well as the human TRPV1 channel. KCNK channels are responsible for the background potassium current of excitable cells, thus essential for the maintenance of the resting membrane potential. Hence, modulators of KCNK channels are of medical significance, e.g. for the treatment of sleep disorders and pain. The transient receptor potential channel TRPV1 is a pain detector for noxious heat. Agonists of this receptor are still used for the treatment of pain in ectopic applications. Experimental procedure We evaluated the effect of 158 herbal remedies on these channels in a heterologous expression system (Xenopus laevis oocytes) using the two-electrode voltage-clamp technique with the aim of increasing the comprehension of their pharmacological profile. Results and conclusion Some remedies with modulating effects were identified such as Angelica pubescens (radix), which inhibit TASK-1 and TASK-3 channels. Furthermore, the modulatory effects of the most effective remedies on the two TASK family members TASK-1 and TASK-3 correlate positively, reflecting their close relation. For the TRPV1 channel Terminalia chebula and Alchemilla xanthochlora were identified as potentiators. This study identifies a variety of herbal remedies as modulators of human K2P and TRPV1 channels and gives new insights into the pharmacological profile of these herbal remedies. Effect of kampo and TCM herbs on human two-pore domain potassium and TRP channels. Effect of 158 herbal remedies on heterologously expressed ion channels. Angelica pubescens (radix) extracts inhibit KCNK3 and KCNK9 channels. Modulatory effects of effective remedies on KCNK3 and KCNK9 correlate positively. Terminalia chebula and Alchemilla xanthochlora are TRPV1 potentiators.
Collapse
Affiliation(s)
- Robin Herbrechter
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | | | - Paul M Ziemba
- AG Physiology of Senses, Ruhr-University Bochum, Bochum, Germany
| | - Sascha Titt
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - Konstantin Lashuk
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - André Gottemeyer
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - Janina Levermann
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - Katrin M Hoffmann
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - Madeline Beltrán
- Department of Receptor Biochemistry, Ruhr-University-Bochum, Bochum, Germany
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - Klemens F Störtkuhl
- Department of Receptor Biochemistry, Ruhr-University-Bochum, Bochum, Germany
| | - Markus Werner
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| | - Günter Gisselmann
- Department of Cell Physiology, Ruhr-University-Bochum, Bochum, Germany
| |
Collapse
|
26
|
Competitive Interactions between Halothane and Isoflurane at the Carotid Body and TASK Channels. Anesthesiology 2020; 133:1046-1059. [DOI: 10.1097/aln.0000000000003520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background
The degree to which different volatile anesthetics depress carotid body hypoxic response relates to their ability to activate TASK potassium channels. Most commonly, volatile anesthetic pairs act additively at their molecular targets. We examined whether this applied to carotid body TASK channels.
Methods
We studied halothane and isoflurane effects on hypoxia-evoked rise in intracellular calcium (Ca2+i, using the indicator Indo-1) in isolated neonatal rat glomus cells, and TASK single-channel activity (patch clamping) in native glomus cells and HEK293 cell line cells transiently expressing TASK-1.
Results
Halothane (5%) depressed glomus cell Ca2+i hypoxic response (mean ± SD, 94 ± 4% depression; P < 0.001 vs. control). Isoflurane (5%) had a less pronounced effect (53 ± 10% depression; P < 0.001 vs. halothane). A mix of 3% isoflurane/1.5% halothane depressed cell Ca2+i response (51 ± 17% depression) to a lesser degree than 1.5% halothane alone (79 ± 15%; P = 0.001), but similar to 3% isoflurane alone (44 ± 22%; P = 0.224), indicating subadditivity. Halothane and isoflurane increased glomus cell TASK-1/TASK-3 activity, but mixes had a lesser effect than that seen with halothane alone: 4% halothane/4% isoflurane yielded channel open probabilities 127 ± 55% above control, versus 226 ± 12% for 4% halothane alone (P = 0.009). Finally, in HEK293 cell line cells, progressively adding isoflurane (1.5 to 5%) to halothane (2.5%) reduced TASK-1 channel activity from 120 ± 38% above control, to 88 ± 48% (P = 0.034).
Conclusions
In all three experimental models, the effects of isoflurane and halothane combinations were quantitatively consistent with the modeling of weak and strong agonists competing at a common receptor on the TASK channel.
Editor’s Perspective
What We Already Know about This Topic
What This Article Tells Us That Is New
Collapse
|
27
|
TASK channels: channelopathies, trafficking, and receptor-mediated inhibition. Pflugers Arch 2020; 472:911-922. [DOI: 10.1007/s00424-020-02403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023]
|
28
|
Ren WJ, Ulrich H, Semyanov A, Illes P, Tang Y. TASK-3: New Target for Pain-Relief. Neurosci Bull 2020; 36:951-954. [PMID: 32458273 DOI: 10.1007/s12264-020-00516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/24/2020] [Indexed: 11/26/2022] Open
Affiliation(s)
- Wen-Jing Ren
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508, Brazil
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997
| | - Peter Illes
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, 04109, Leipzig, Sachsen, Germany.
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
29
|
Wiedmann F, Beyersdorf C, Zhou X, Büscher A, Kraft M, Nietfeld J, Walz TP, Unger LA, Loewe A, Schmack B, Ruhparwar A, Karck M, Thomas D, Borggrefe M, Seemann G, Katus HA, Schmidt C. Pharmacologic TWIK-Related Acid-Sensitive K+ Channel (TASK-1) Potassium Channel Inhibitor A293 Facilitates Acute Cardioversion of Paroxysmal Atrial Fibrillation in a Porcine Large Animal Model. J Am Heart Assoc 2020; 9:e015751. [PMID: 32390491 PMCID: PMC7660874 DOI: 10.1161/jaha.119.015751] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
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.
Collapse
Affiliation(s)
- Felix Wiedmann
- Department of Cardiology University of Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Christoph Beyersdorf
- Department of Cardiology University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Xiaobo Zhou
- DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,First Department of Medicine University Medical Center Mannheim Germany
| | - Antonius Büscher
- Department of Cardiology University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Manuel Kraft
- Department of Cardiology University of Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Jendrik Nietfeld
- Department of Cardiology University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Teo Puig Walz
- Institute for Experimental Cardiovascular Medicine University Heart Center Freiburg Bad Krozingen Germany.,Medical Center University of Freiburg, and Faculty of Medicine University of Freiburg Germany
| | - Laura A Unger
- Institute of Biomedical Engineering Karlsruhe Institute of Technology (KIT) Karlsruhe Germany
| | - Axel Loewe
- Institute of Biomedical Engineering Karlsruhe Institute of Technology (KIT) Karlsruhe Germany
| | - Bastian Schmack
- Department of Cardiac Surgery University Hospital Heidelberg Germany
| | | | - Matthias Karck
- Department of Cardiac Surgery University Hospital Heidelberg Germany
| | - Dierk Thomas
- Department of Cardiology University of Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Martin Borggrefe
- DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,First Department of Medicine University Medical Center Mannheim Germany
| | - Gunnar Seemann
- Institute for Experimental Cardiovascular Medicine University Heart Center Freiburg Bad Krozingen Germany.,Medical Center University of Freiburg, and Faculty of Medicine University of Freiburg Germany
| | - Hugo A Katus
- Department of Cardiology University of Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| | - Constanze Schmidt
- Department of Cardiology University of Heidelberg Germany.,DZHK (German Center for Cardiovascular Research) partner site Heidelberg /Mannheim University of Heidelberg Germany.,HCR Heidelberg Center for Heart Rhythm Disorders University of Heidelberg Germany
| |
Collapse
|
30
|
Busserolles J, Lolignier S, Kerckhove N, Bertin C, Authier N, Eschalier A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol Ther 2020; 210:107519. [PMID: 32165137 DOI: 10.1016/j.pharmthera.2020.107519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
The scarcity and limited risk/benefit ratio of painkillers available on the market, in addition to the opioid crisis, warrant reflection on new innovation strategies. The pharmacopoeia of analgesics is based on products that are often old and derived from clinical empiricism, with limited efficacy or spectrum of action, or resulting in an unsatisfactory tolerability profile. Although they are reference analgesics for nociceptive pain, opioids are subject to the same criticism. The use of opium as an analgesic is historical. Morphine was synthesized at the beginning of the 19th century. The efficacy of opioids is limited in certain painful contexts and these drugs can induce potentially serious and fatal adverse effects. The current North American opioid crisis, with an ever-rising number of deaths by opioid overdose, is a tragic illustration of this. It is therefore legitimate to develop research into molecules likely to maintain or increase opioid efficacy while improving their tolerability. Several avenues are being explored including targeting of the mu opioid receptor (MOR) splice variants, developing biased agonists or targeting of other receptors such as heteromers with MOR. Ion channels acting as MOR effectors, are also targeted in order to offer compounds without MOR-dependent adverse effects. Another route is to develop opioid analgesics with peripheral action or limited central nervous system (CNS) access. Finally, endogenous opioids used as drugs or compounds that modify the metabolism of endogenous opioids (Dual ENKephalinase Inhibitors) are being developed. The aim of the present review is to present these various targets/strategies with reference to current indications for opioids, concerns about their widespread use, particularly in chronic non-cancer pains, and ways of limiting the risk of opioid abuse and misuse.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Nicolas Kerckhove
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Célian Bertin
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
31
|
Zhou T, Luo P, Wang L, Yang S, Qin S, Wei Z, Liu J. CTNNB1 Knockdown Inhibits Cell Proliferation and Aldosterone Secretion Through Inhibiting Wnt/β-Catenin Signaling in H295R Cells. Technol Cancer Res Treat 2020; 19:1533033820979685. [PMID: 33287648 PMCID: PMC7727057 DOI: 10.1177/1533033820979685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/09/2020] [Accepted: 11/02/2020] [Indexed: 01/31/2023] Open
Abstract
Aldosterone-producing adenomas (APA) is one of the causative factors of primary aldosteronism. Previous studies have suggested that there are somatic CTNNB1 mutations in APA, but the specific mechanism of CTNNB1 mutation in APA tumorigenesis and aldosterone secretion remains unclear. In the present study, human adrenocortical carcinoma cell line H295 R was used to establish stable CTNNB1 knockdown cell lines. Cell proliferation and aldosterone secretion of H295 R cells in response to angiotensin Ⅱ (Agn Ⅱ) were analyzed. We found that CTNNB1 knockdown reduced β-catenin expression and inhibited proliferation of H295 R cells. CTNNB1 knockdown inhibited Wnt/β-catenin signaling pathway and downregulated expression of downstream genes including axin 2, lymphoid enhancer binding factor 1 (LEF1), and cyclin D1. In addition, CTNNB1 knockdown decreased responses of H295 R cells to Agn Ⅱ and decreased aldosterone secretion. Our findings suggest that CTNNB1 knockdown can inhibit H295 R cell proliferation and decrease aldosterone secretion in the responses of H295 R cells to Ang II through inhibiting Wnt/β-catenin signaling pathway, indicating that targeting Wnt/β-catenin signaling pathway may be an important approach to decrease aldosterone secretion in the treatment of aldoster-producing adenomas.
Collapse
Affiliation(s)
- Tingting Zhou
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Pengwei Luo
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
- Department of Urology, The First Affiliated Hospital of Chengdu Medical College
| | - Liang Wang
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Shiwei Yang
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Shiyuan Qin
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Zhitao Wei
- Department of Urology, The First Affiliated Hospital of Chengdu Medical College
| | - Jiwen Liu
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| |
Collapse
|
32
|
Muscarinic receptor stimulation induces TASK1 channel endocytosis through a PKC-Pyk2-Src pathway in PC12 cells. Cell Signal 2020; 65:109434. [DOI: 10.1016/j.cellsig.2019.109434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 11/21/2022]
|
33
|
Guagliardo NA, Yao J, Stipes EJ, Cechova S, Le TH, Bayliss DA, Breault DT, Barrett PQ. Adrenal Tissue-Specific Deletion of TASK Channels Causes Aldosterone-Driven Angiotensin II-Independent Hypertension. Hypertension 2019; 73:407-414. [PMID: 30580687 PMCID: PMC6326871 DOI: 10.1161/hypertensionaha.118.11962] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The renin-angiotensin system tightly controls aldosterone synthesis. Dysregulation is evident in hypertension (primary aldosteronism), low renin, and resistant hypertension) but also can exist in normotension. Whether chronic, mild aldosterone autonomy can elicit hypertension remains untested. Previously, we reported that global genetic deletion of 2 pore-domain TWIK-relative acid-sensitive potassium channels, TASK-1 and TASK-3, from mice produces striking aldosterone excess, low renin, and hypertension. Here, we deleted TASK-1 and TASK-3 channels selectively from zona glomerulosa cells and generated a model of mild aldosterone autonomy with attendant hypertension that is aldosterone-driven and Ang II (angiotensin II)-independent. This study shows that a zona glomerulosa-specific channel defect can produce mild autonomous hyperaldosteronism sufficient to cause chronic blood pressure elevation.
Collapse
Affiliation(s)
- Nick A Guagliardo
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - Junlan Yao
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - Eric J Stipes
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - Sylvia Cechova
- Division of Nephrology, Department of Medicine (S.C., T.H.L.), University of Virginia School of Medicine, Charlottesville
| | - Thu H Le
- Division of Nephrology, Department of Medicine (S.C., T.H.L.), University of Virginia School of Medicine, Charlottesville
| | - Douglas A Bayliss
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| | - David T Breault
- Department of Pediatrics/Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, MA (D.T.B.)
| | - Paula Q Barrett
- From the Department of Pharmacology (N.A.G., J.Y., E.J.S., D.A.B, P.Q.B.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
34
|
García G, Noriega-Navarro R, Martínez-Rojas VA, Gutiérrez-Lara EJ, Oviedo N, Murbartián J. Spinal TASK-1 and TASK-3 modulate inflammatory and neuropathic pain. Eur J Pharmacol 2019; 862:172631. [DOI: 10.1016/j.ejphar.2019.172631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/09/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
|
35
|
Sp1-regulated expression of p11 contributes to motor neuron degeneration by membrane insertion of TASK1. Nat Commun 2019; 10:3784. [PMID: 31439839 PMCID: PMC6706379 DOI: 10.1038/s41467-019-11637-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 07/25/2019] [Indexed: 01/18/2023] Open
Abstract
Disruption in membrane excitability contributes to malfunction and differential vulnerability of specific neuronal subpopulations in a number of neurological diseases. The adaptor protein p11, and background potassium channel TASK1, have overlapping distributions in the CNS. Here, we report that the transcription factor Sp1 controls p11 expression, which impacts on excitability by hampering functional expression of TASK1. In the SOD1-G93A mouse model of ALS, Sp1-p11-TASK1 dysregulation contributes to increased excitability and vulnerability of motor neurons. Interference with either Sp1 or p11 is neuroprotective, delaying neuron loss and prolonging lifespan in this model. Nitrosative stress, a potential factor in human neurodegeneration, stimulated Sp1 expression and human p11 promoter activity, at least in part, through a Sp1-binding site. Disruption of Sp1 or p11 also has neuroprotective effects in a traumatic model of motor neuron degeneration. Together our work suggests the Sp1-p11-TASK1 pathway is a potential target for treatment of degeneration of motor neurons. The adaptor protein p11 and K+ channel TASK1 have overlapping distributions in the CNS. Here, the authors demonstrate that the transcription factor Sp1 regulates p11 levels, which in turn affects intrinsic membrane properties and can contribute to degeneration of motor neurons in disease and injury models.
Collapse
|
36
|
Discovery of Novel TASK-3 Channel Blockers Using a Pharmacophore-Based Virtual Screening. Int J Mol Sci 2019; 20:ijms20164014. [PMID: 31426491 PMCID: PMC6720600 DOI: 10.3390/ijms20164014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
TASK-3 is a two-pore domain potassium (K2P) channel highly expressed in the hippocampus, cerebellum, and cortex. TASK-3 has been identified as an oncogenic potassium channel and it is overexpressed in different cancer types. For this reason, the development of new TASK-3 blockers could influence the pharmacological treatment of cancer and several neurological conditions. In the present work, we searched for novel TASK-3 blockers by using a virtual screening protocol that includes pharmacophore modeling, molecular docking, and free energy calculations. With this protocol, 19 potential TASK-3 blockers were identified. These molecules were tested in TASK-3 using patch clamp, and one blocker (DR16) was identified with an IC50 = 56.8 ± 3.9 μM. Using DR16 as a scaffold, we designed DR16.1, a novel TASK-3 inhibitor, with an IC50 = 14.2 ± 3.4 μM. Our finding takes on greater relevance considering that not many inhibitory TASK-3 modulators have been reported in the scientific literature until today. These two novel TASK-3 channel inhibitors (DR16 and DR16.1) are the first compounds found using a pharmacophore-based virtual screening and rational drug design protocol.
Collapse
|
37
|
Vyas VK, Parikh P, Ramani J, Ghate M. Medicinal Chemistry of Potassium Channel Modulators: An Update of Recent Progress (2011-2017). Curr Med Chem 2019; 26:2062-2084. [PMID: 29714134 DOI: 10.2174/0929867325666180430152023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 12/22/2017] [Accepted: 04/25/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Potassium (K+) channels participate in many physiological processes, cardiac function, cell proliferation, neuronal signaling, muscle contractility, immune function, hormone secretion, osmotic pressure, changes in gene expression, and are involved in critical biological functions, and in a variety of diseases. Potassium channels represent a large family of tetrameric membrane proteins. Potassium channels activation reduces excitability, whereas channel inhibition increases excitability. OBJECTIVE Small molecule K+ channel activators and inhibitors interact with voltage-gated, inward rectifying, and two-pore tandem potassium channels. Due to their involvement in biological functions, and in a variety of diseases, small molecules as potassium channel modulators have received great scientific attention. METHODS In this review, we have compiled the literature, patents and patent applications (2011 to 2017) related to different chemical classes of potassium channel openers and blockers as therapeutic agents for the treatment of various diseases. Many different chemical classes of selective small molecule have emerged as potassium channel modulators over the past years. CONCLUSION This review discussed the current understanding of medicinal chemistry research in the field of potassium channel modulators to update the key advances in this field.
Collapse
Affiliation(s)
- Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481 Gujarat, India
| | - Palak Parikh
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481 Gujarat, India
| | - Jonali Ramani
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481 Gujarat, India
| | - Manjunath Ghate
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382 481 Gujarat, India
| |
Collapse
|
38
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
39
|
Pineda RH, Hypolite J, Lee S, Carrasco A, Iguchi N, Meacham RB, Malykhina AP. Altered detrusor contractility and voiding patterns in mice lacking the mechanosensitive TREK-1 channel. BMC Urol 2019; 19:40. [PMID: 31113422 PMCID: PMC6528348 DOI: 10.1186/s12894-019-0475-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/13/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Previously published results from our laboratory identified a mechano-gated two-pore domain potassium channel, TREK-1, as a main mechanosensor in the smooth muscle of the human urinary bladder. One of the limitations of in vitro experiments on isolated human detrusor included inability to evaluate in vivo effects of TREK-1 on voiding function, as the channel is also expressed in the nervous system, and may modulate micturition via neural pathways. Therefore, in the present study, we aimed to assess the role of TREK-1 channel in bladder function and voiding patterns in vivo by using TREK-1 knockout (KO) mice. METHODS Adult C57BL/6 J wild-type (WT, N = 32) and TREK-1 KO (N = 33) mice were used in this study. The overall phenotype and bladder function were evaluated by gene and protein expression of TREK-1 channel, in vitro contractile experiments using detrusor strips in response to stretch and pharmacological stimuli, and cystometry in unanesthetized animals. RESULTS TREK-1 KO animals had an elevated basal muscle tone and enhanced spontaneous activity in the detrusor without detectable changes in bladder morphology/histology. Stretch applied to isolated detrusor strips increased the amplitude of spontaneous contractions by 109% in the TREK-1 KO group in contrast to a 61% increase in WT mice (p ≤ 0.05 to respective baseline for each group). The detrusor strips from TREK-1 KO mice also generated more contractile force in response to electric field stimulation and high potassium concentration in comparison to WT group (p ≤ 0.05 for both tests). However, cystometric recordings from TREK-1 KO mice revealed a significant increase in the duration of the intermicturition interval, enhanced bladder capacity and increased number of non-voiding contractions in comparison to WT mice. CONCLUSIONS Our results provide evidence that global down-regulation of TREK-1 channels has dual effects on detrusor contractility and micturition patterns in vivo. The observed differences are likely due to expression of TREK-1 channel not only in detrusor myocytes but also in afferent and efferent neural pathways involved in regulation of micturition which may underly the "mixed" voiding phenotype in TREK-1 KO mice.
Collapse
Affiliation(s)
- Ricardo H Pineda
- Division of Urology, Department of Surgery, University of Colorado Denver,Anschutz Medical Campus, 12700 E 19th Ave, M/S C317, Aurora, CO, 80045, USA
| | - Joseph Hypolite
- Division of Urology, Department of Surgery, University of Colorado Denver,Anschutz Medical Campus, 12700 E 19th Ave, M/S C317, Aurora, CO, 80045, USA
| | - Sanghee Lee
- Department of Urology, University of California San Diego, 3855 Health Science Drive, Room 4345, Bay 4LL, La Jolla, CA, 92093, USA
| | - Alonso Carrasco
- Children's Mercy Hospital, 2401 Gillham Rd, Kansas City, MO, 64108, USA
| | - Nao Iguchi
- Division of Urology, Department of Surgery, University of Colorado Denver,Anschutz Medical Campus, 12700 E 19th Ave, M/S C317, Aurora, CO, 80045, USA
| | - Randall B Meacham
- Division of Urology, Department of Surgery, University of Colorado Denver, Academic Office One Bldg., Rm 5602, 12631 East 17th Ave., M/S C319, Aurora, CO, 80045, USA
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver,Anschutz Medical Campus, 12700 E 19th Ave, M/S C317, Aurora, CO, 80045, USA.
| |
Collapse
|
40
|
Lamas JA, Rueda-Ruzafa L, Herrera-Pérez S. Ion Channels and Thermosensitivity: TRP, TREK, or Both? Int J Mol Sci 2019; 20:ijms20102371. [PMID: 31091651 PMCID: PMC6566417 DOI: 10.3390/ijms20102371] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 11/16/2022] Open
Abstract
Controlling body temperature is a matter of life or death for most animals, and in mammals the complex thermoregulatory system is comprised of thermoreceptors, thermosensors, and effectors. The activity of thermoreceptors and thermoeffectors has been studied for many years, yet only recently have we begun to obtain a clear picture of the thermosensors and the molecular mechanisms involved in thermosensory reception. An important step in this direction was the discovery of the thermosensitive transient receptor potential (TRP) cationic channels, some of which are activated by increases in temperature and others by a drop in temperature, potentially converting the cells in which they are expressed into heat and cold receptors. More recently, the TWIK-related potassium (TREK) channels were seen to be strongly activated by increases in temperature. Hence, in this review we want to assess the hypothesis that both these groups of channels can collaborate, possibly along with other channels, to generate the wide range of thermal sensations that the nervous system is capable of handling.
Collapse
Affiliation(s)
- J Antonio Lamas
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, 36310 Vigo, Spain.
| | - Lola Rueda-Ruzafa
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, 36310 Vigo, Spain.
| | - Salvador Herrera-Pérez
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, 36310 Vigo, Spain.
| |
Collapse
|
41
|
García G, Gutiérrez-Lara EJ, Centurión D, Granados-Soto V, Murbartián J. Fructose-Induced Insulin Resistance as a Model of Neuropathic Pain in Rats. Neuroscience 2019; 404:233-245. [DOI: 10.1016/j.neuroscience.2019.01.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/21/2022]
|
42
|
Kawasaki K, Suzuki Y, Yamamura H, Imaizumi Y. Development of a Novel Cell-Based Assay System for High-Throughput Screening of Compounds Acting on Background Two-Pore Domain K + Channels. SLAS DISCOVERY 2019; 24:641-652. [PMID: 30802418 DOI: 10.1177/2472555219829745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Two-pore domain K+ (K2P) channels are thought to be druggable targets. However, only a few agents specific for K2P channels have been identified, presumably due to the lack of an efficient screening system. To develop a new high-throughput screening (HTS) system targeting these channels, we have established a HEK293-based "test cell" expressing a mutated Na+ channel (Nav1.5) with markedly slowed inactivation, as well as a K+ channel (Kir2.1) that sets the membrane potential quite negative, close to K+ equilibrium potential. We found in this system that Kir2.1 block by 100 μM Ba2+ application consistently elicited a large depolarization like a long-lasting action potential. This maneuver resulted in cell death, presumably due to the sustained Na+ influx. When either the TWIK-related acid-sensitive K+ (TASK)-1 or TASK-3 channel was expressed in the test cells, Ba2+-induced cell death was markedly weakened. Stronger activation of TASK-1 by extracellular acidification further decreased the cell death. In contrast, the presence of K2P channel blockers enhanced cell death. IC50 values for TASK-1 and/or TASK-3 blockers acquired by measurements of relative cell viability were comparable to those obtained using patch-clamp recordings. Both blockers and openers of K2P channels can be accurately assessed with high efficiency and throughput by this novel HTS system.
Collapse
Affiliation(s)
- Keisuke Kawasaki
- 1 Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yoshiaki Suzuki
- 1 Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Hisao Yamamura
- 1 Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yuji Imaizumi
- 1 Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.,2 Department of Research and Development, ChanneloSearch Technology Co., Ltd., Nagoya, Japan
| |
Collapse
|
43
|
Canella R, Martini M, Cavicchio C, Cervellati F, Benedusi M, Valacchi G. Involvement of the TREK-1 channel in human alveolar cell membrane potential and its regulation by inhibitors of the chloride current. J Cell Physiol 2019; 234:17704-17713. [PMID: 30805940 DOI: 10.1002/jcp.28396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
K+ channels of the alveolar epithelium control the driving force acting on the ionic and solvent flow through the cell membrane contributing to the maintenance of cell volume and the constitution of epithelial lining fluid. In the present work, we analyze the effect of the Cl- channel inhibitors: (4-[(2-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-inden-5-yl)oxy] butanoic acid (DCPIB) and 9-anthracenecarboxylic acid (9-AC) on the total current in a type II pneumocytes (A549 cell line) model by patch clamp, immunocytochemical, and gene knockdown techniques. We noted that DCPIB and 9-AC promote the activation of K conductance. In fact, they significantly increase the intensity of the current and shift its reversal potential to values more negative than the control. By silencing outward rectifier channel in its anoctamin 6 portion, we excluded a direct involvement of Cl- ions in modulation of IK and, by means of functional tests with its specific inhibitor spadin, we identified the TREK-1 channel as the presumable target of both drugs. As the activity of TREK-1 has a key role for the correct functioning of the alveolar epithelium, the identification of DCPIB and 9-AC molecules as its activators suggests their possible use to build new pharmacological tools for the modulation of this channel.
Collapse
Affiliation(s)
- Rita Canella
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Marta Martini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Carlotta Cavicchio
- Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina
| | - Franco Cervellati
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mascia Benedusi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina
| |
Collapse
|
44
|
Zavala WD, Foscolo MR, Kunda PE, Cavicchia JC, Acosta CG. Changes in the expression of the potassium channels TASK1, TASK3 and TRESK in a rat model of oral squamous cell carcinoma and their relation to malignancy. Arch Oral Biol 2019; 100:75-85. [PMID: 30818127 DOI: 10.1016/j.archoralbio.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Potassium channels have been proposed to promote cancer cell proliferation and metastases. Thus, we investigated the expression pattern of three 2-pore domain potassium channels (K2Ps) TASK1, TASK3 and TRESK in advanced oral squamous cell carcinoma (OSCC), the commonest oral malignancy. DESIGN We used 4-nitroquinoline-1-oxide (4-NQO) to induce high grade OSCC in male adult rats. We then used immunohistochemistry and Western blotting to study the distribution and expression pattern of TASK1, TASK3 and TRESK in normal versus cancerous tissue. We also examined the expression of β-tubulin III (β-tub3), a marker associated with resistance to taxane-based chemotherapy and poor patient prognosis, and its correlation with the K2Ps. Finally, we studied the expression of TASK1, TASK3 and TRESK in human samples of SCC of oral origin. RESULTS We found that TASK3 was significantly up-regulated whereas TASK1 and TRESK were both significantly down-regulated in advanced, poorly differentiated OSCC. Both, rat and human SCC showed a significant increase in the expression of β-tub3. Interestingly, the expression of the latter correlated positively and significantly with TASK3 and TRESK but not TASK1 in rat OSCC. Our initial results showed a similar pattern of up and down regulation and correlation with β-tub3 for these three K2Ps in human SCC. CONCLUSIONS The changes in expression and the co-localization with a marker of resistance to taxanes like β-tub3 turn TASK1, TASK3 and TRESK into potentially new prognostic tools and possibly new therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Walther D Zavala
- Facultad de Odontología, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Mabel R Foscolo
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Patricia E Kunda
- Centro Investigación Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina.
| | - Juan C Cavicchia
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Cristian G Acosta
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
45
|
Inoue M, Harada K, Matsui M, Matsuoka H. Differences among muscarinic agonists in M 1 receptor-mediated nonselective cation channel activation and TASK1 channel inhibition in adrenal medullary cells. Eur J Pharmacol 2019; 843:104-112. [PMID: 30452911 DOI: 10.1016/j.ejphar.2018.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 10/27/2022]
Abstract
Muscarinic receptor stimulation induces depolarizing inward currents and catecholamine secretion in adrenal medullary (AM) cells from various mammals. In guinea-pig AM cells muscarine and oxotremorine at concentrations ≤ 1 μM produce activation of nonselective cation channels with a similar potency and efficacy, whereas muscarine at higher concentrations produces not only nonselective cation channel activation, but also TASK1 channel inhibition. In rat AM cells, the muscarinic M1 receptor is involved in TASK1 channel inhibition in response to muscarinic agonists, and the efficacy of oxotremorine is half that of muscarine. These pharmacological findings might indicate that different muscarinic receptor subtypes are responsible for the regulation of nonselective cation and TASK1 channel activities. The present study aimed to determine the muscarinic receptor subtypes involved in nonselective cation channel activation in guinea-pig and mouse AM cells. The inward current evoked by 1 μM muscarine was completely suppressed by 100 μM quinine, whereas 30 μM muscarine-induced inward currents were comprised of quinine-sensitive and -insensitive components. The electrophysiological and pharmacological properties of the muscarine-induced currents indicated that the quinine-sensitive and insensitive components are due to nonselective cation channel activation and TASK1 channel inhibition, respectively. Muscarine at 30 μM failed to induce any current in AM cells treated with muscarinic toxin 7 or genetically deleted of the M1 receptor. The KD value of VU0255035 against the muscarinic receptor mediating nonselective cation channel activation was 17.5 nM. These results indicate that the M1 receptor mediates nonselective cation channel activation as well as TASK1 channel inhibition.
Collapse
Affiliation(s)
- Masumi Inoue
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan.
| | - Keita Harada
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | | | - Hidetada Matsuoka
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| |
Collapse
|
46
|
Lamas JA, Fernández-Fernández D. Tandem pore TWIK-related potassium channels and neuroprotection. Neural Regen Res 2019; 14:1293-1308. [PMID: 30964046 PMCID: PMC6524494 DOI: 10.4103/1673-5374.253506] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
TWIK-related potassium channels (TREK) belong to a subfamily of the two-pore domain potassium channels family with three members, TREK1, TREK2 and TWIK-related arachidonic acid-activated potassium channels. The two-pore domain potassium channels is the last big family of channels being discovered, therefore it is not surprising that most of the information we know about TREK channels predominantly comes from the study of heterologously expressed channels. Notwithstanding, in this review we pay special attention to the limited amount of information available on native TREK-like channels and real neurons in relation to neuroprotection. Mainly we focus on the role of free fatty acids, lysophospholipids and other neuroprotective agents like riluzole in the modulation of TREK channels, emphasizing on how important this modulation may be for the development of new therapies against neuropathic pain, depression, schizophrenia, epilepsy, ischemia and cardiac complications.
Collapse
Affiliation(s)
- J Antonio Lamas
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| | - Diego Fernández-Fernández
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| |
Collapse
|
47
|
Pope L, Arrigoni C, Lou H, Bryant C, Gallardo-Godoy A, Renslo AR, Minor DL. Protein and Chemical Determinants of BL-1249 Action and Selectivity for K 2P Channels. ACS Chem Neurosci 2018; 9:3153-3165. [PMID: 30089357 PMCID: PMC6302903 DOI: 10.1021/acschemneuro.8b00337] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
K2P potassium channels generate leak currents that stabilize the resting membrane potential of excitable cells. Various K2P channels are implicated in pain, ischemia, depression, migraine, and anesthetic responses, making this family an attractive target for small molecule modulator development efforts. BL-1249, a compound from the fenamate class of nonsteroidal anti-inflammatory drugs is known to activate K2P2.1(TREK-1), the founding member of the thermo- and mechanosensitive TREK subfamily; however, its mechanism of action and effects on other K2P channels are not well-defined. Here, we demonstrate that BL-1249 extracellular application activates all TREK subfamily members but has no effect on other K2P subfamilies. Patch clamp experiments demonstrate that, similar to the diverse range of other chemical and physical TREK subfamily gating cues, BL-1249 stimulates the selectivity filter "C-type" gate that controls K2P function. BL-1249 displays selectivity among the TREK subfamily, activating K2P2.1(TREK-1) and K2P10.1(TREK-2) ∼10-fold more potently than K2P4.1(TRAAK). Investigation of mutants and K2P2.1(TREK-1)/K2P4.1(TRAAK) chimeras highlight the key roles of the C-terminal tail in BL-1249 action and identify the M2/M3 transmembrane helix interface as a key site of BL-1249 selectivity. Synthesis and characterization of a set of BL-1249 analogs demonstrates that both the tetrazole and opposing tetralin moieties are critical for function, whereas the conformational mobility between the two ring systems impacts selectivity. Together, our findings underscore the landscape of modes by which small molecules can affect K2P channels and provide crucial information for the development of better and more selective K2P modulators of the TREK subfamily.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniel L. Minor
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 United States
| |
Collapse
|
48
|
Seccia TM, Caroccia B, Gomez-Sanchez EP, Gomez-Sanchez CE, Rossi GP. The Biology of Normal Zona Glomerulosa and Aldosterone-Producing Adenoma: Pathological Implications. Endocr Rev 2018; 39:1029-1056. [PMID: 30007283 PMCID: PMC6236434 DOI: 10.1210/er.2018-00060] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
The identification of several germline and somatic ion channel mutations in aldosterone-producing adenomas (APAs) and detection of cell clusters that can be responsible for excess aldosterone production, as well as the isolation of autoantibodies activating the angiotensin II type 1 receptor, have rapidly advanced the understanding of the biology of primary aldosteronism (PA), particularly that of APA. Hence, the main purpose of this review is to discuss how discoveries of the last decade could affect histopathology analysis and clinical practice. The structural remodeling through development and aging of the human adrenal cortex, particularly of the zona glomerulosa, and the complex regulation of aldosterone, with emphasis on the concepts of zonation and channelopathies, will be addressed. Finally, the diagnostic workup for PA and its subtyping to optimize treatment are reviewed.
Collapse
Affiliation(s)
- Teresa M Seccia
- Department of Medicine-DIMED, University of Padua, Padua PD, Italy
| | | | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi
| | - Celso E Gomez-Sanchez
- Division of Endocrinology, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi.,University of Mississippi Medical Center, Jackson, Mississippi
| | - Gian Paolo Rossi
- Department of Medicine-DIMED, University of Padua, Padua PD, Italy
| |
Collapse
|
49
|
Wang X, Guan R, Zhao X, Zhu D, Song N, Shen L. TASK1 and TASK3 Are Coexpressed With ASIC1 in the Ventrolateral Medulla and Contribute to Central Chemoreception in Rats. Front Cell Neurosci 2018; 12:285. [PMID: 30210304 PMCID: PMC6123564 DOI: 10.3389/fncel.2018.00285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/10/2018] [Indexed: 11/13/2022] Open
Abstract
The ventrolateral medulla (VLM), including the lateral paragigantocellular nucleus (LPGi) and rostral VLM (RVLM), is commonly considered to be a chemosensitive region. However, the specific mechanism of chemoreception in the VLM remains elusive. Acid-sensing ion channels (ASICs), a family of voltage-independent proton-gated cation channels, can be activated by an external pH decrease to cause Na+ entry and induce neuronal excitability. TWIK-related acid-sensitive potassium channels (TASKs) are members of another group of pH-sensitive channels; in contrast to AISICs, they can be stimulated by pH increases and are inhibited by pH decreases in the physiological range. Our previous study demonstrated that ASICs take part in chemoreception. The aims of this study are to explore whether TASKs participate in the acid sensitivity of neurons in the VLM, thereby cooperating with ASICs. Our research demonstrated that TASKs, including TASK1 and TASK3, are colocalized with ASIC1 in VLM neurons. Blocking TASKs by microinjection of the non-selective TASK antagonist bupivacaine (BUP), specific TASK1 antagonist anandamide (AEA) or specific TASK3 antagonist ruthenium red (RR) into the VLM increased the integrated phrenic nerve discharge (iPND), shortened the inspiratory time (Ti) and enhanced the respiratory drive (iPND/Ti). In addition, microinjection of artificial cerebrospinal fluid (ACSF) at a pH of 7.0 or 6.5 prolonged Ti, increased iPND and enhanced respiratory drive, which were inhibited by the ASIC antagonist amiloride (AMI). By contrast, microinjection of alkaline ACSF decreased iPND and respiratory drive, which were inhibited by AEA. Taken together, our data suggest that TASK1 and TASK3 are coexpressed with ASIC1 in the VLM. Moreover, TASK1 and TASK3 contribute to the central regulation of breathing by coordinating with each other to perceive local pH changes; these results indicate a novel chemosensitive mechanism of the VLM.
Collapse
Affiliation(s)
- Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ruijuan Guan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaomei Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Danian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nana Song
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linlin Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Medical Imaging Computing and Computer-Assisted Intervention, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Huang H, Li H, Shi K, Wang L, Zhang X, Zhu X. TREK‑TRAAK two‑pore domain potassium channels protect human retinal pigment epithelium cells from oxidative stress. Int J Mol Med 2018; 42:2584-2594. [PMID: 30106090 PMCID: PMC6192786 DOI: 10.3892/ijmm.2018.3813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 07/31/2018] [Indexed: 12/19/2022] Open
Abstract
The aim of the current study was to explore the potential of TREK‑TRAAK two‑pore domain potassium (K2P) channels in protecting human retinal pigment epithelium (hRPE) cells against oxidative stress. hRPE cells were obtained from donors, and then cell identification and detection of the expression levels of TREK‑TRAAK K2P channels in hRPE cells were conducted. Subsequently, tert‑butyl hydroperoxide (t‑BH) was used to induce oxidative stress in hRPE cells. Docosahexaenoic acid (DHA) was used to stimulate and fluoxetine was used to inhibit the TREK‑TRAAK K2P channels. The survival rates of hRPE cells under oxidative stress were examined using flow cytometry. Apoptosis‑associated factors, including Bax, Bcl‑2, cleaved‑caspase‑3, αB‑crystallin and their mRNAs, were examined using immunofluorescence, western blot and reverse transcription‑polymerase chain reaction analyses. Variations in the cytoarchitecture were observed by immunofluorescence and electron microscopy. The cells examined in the present study were identified as hRPE cells. All members in the TREK‑TRAAK K2P channel family (including TREK‑1, TREK‑2 and TRAAK) were found to be expressed in hRPE cells. Stimulation of TREK‑TRAAK K2P channels increased the survival rates of hRPE cells under oxidative stress and the levels of intracellular protective factors, such as Bcl‑2 and αB‑crystallin. By contrast, inhibition of these channels decreased the cell survival rates and increased apoptosis enhancing factors, such as Bax and cleaved‑caspase‑3. Further examination of the cytoarchitecture revealed that TREK‑TRAAK K2P channels protected the integrity of the hRPE cell structure against oxidative stress. In conclusion, the present study suggested that the activated TREK‑TRAAK K2P channels serve a role in protecting hRPE cells against the oxidative stress induced by t‑BH, which indicated that these K2P channels are potential novel targets in retinal protection and provided a new direction for research and therapy in retinal degeneration diseases.
Collapse
Affiliation(s)
- Hao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Han Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Kangpei Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Lei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaotong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaobo Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|