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
|
Liu C, Chen IS, Tateyama M, Kubo Y. Structural determinants of the direct inhibition of GIRK channels by Sigma-1 receptor antagonist. J Biol Chem 2024; 300:107219. [PMID: 38522516 PMCID: PMC11031820 DOI: 10.1016/j.jbc.2024.107219] [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: 11/22/2023] [Revised: 03/05/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
G-protein-gated inward rectifier K+ (GIRK) channels play a critical role in the regulation of the excitability of cardiomyocytes and neurons and include GIRK1, GIRK2, GIRK3 and GIRK4 subfamily members. BD1047 dihydrobromide (BD1047) is one of the representative antagonists of the multifunctional Sigma-1 receptor (S1R). In the analysis of the effect of BD1047 on the regulation of Gi-coupled receptors by S1R using GIRK channel as an effector, we observed that BD1047, as well as BD1063, directly inhibited GIRK currents even in the absence of S1R and in a voltage-independent manner. Thus, we aimed to clarify the effect of BD1047 on GIRK channels and identify the structural determinants. By electrophysiological recordings in Xenopus oocytes, we observed that BD1047 directly inhibited GIRK channel currents, producing a much stronger inhibition of GIRK4 compared to GIRK2. It also inhibited ACh-induced native GIRK current in isolated rat atrial myocytes. Chimeric and mutagenesis studies of GIRK2 and GIRK4 combined with molecular docking analysis demonstrated the importance of Leu77 and Leu84 within the cytoplasmic, proximal N-terminal region and Glu147 within the pore-forming region of GIRK4 for inhibition by BD1047. The activator of GIRK channels, ivermectin, competed with BD1047 at Leu77 on GIRK4. This study provides us with a novel inhibitor of GIRK channels and information for developing pharmacological treatments for GIRK4-associated diseases.
Collapse
Affiliation(s)
- Chang Liu
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan.
| | - I-Shan Chen
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan; Faculty of Medicine, Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Michihiro Tateyama
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan.
| |
Collapse
|
3
|
GIRK Channels as Candidate Targets for the Treatment of Substance Use Disorders. Biomedicines 2022; 10:biomedicines10102552. [PMID: 36289814 PMCID: PMC9599444 DOI: 10.3390/biomedicines10102552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/05/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
Substance use disorders (SUDs) are chronic, lifelong disorders that have serious consequences. Repeated substance use alters brain function. G-protein-activated inwardly rectifying potassium (GIRK) channels are expressed widely in the brain, including the reward system, and regulate neuronal excitability. Functional GIRK channels are identified as heterotetramers of GIRK subunits (GIRK1–4). The GIRK1, GIRK2, and GIRK3 subunits are mainly expressed in rodent brain regions, and various addictive substances act on the brain through GIRK channels. Studies with animals (knockout and missense mutation animals) and humans have demonstrated the involvement of GIRK channels in the effects of addictive substances. Additionally, GIRK channel blockers affect behavioral responses to addictive substances. Thus, GIRK channels play a key role in SUDs, and GIRK channel modulators may be candidate medications. Ifenprodil is a GIRK channel blocker that does not have serious side effects. Two clinical trials were conducted to investigate the effects of ifenprodil in patients with alcohol or methamphetamine use disorder. Although the number of participants was relatively low, evidence of its safety and efficacy was found. The present review discusses the potential of GIRK channel modulators as possible medications for addiction. Therapeutic agents that target GIRK channels may be promising for the treatment of SUDs.
Collapse
|
4
|
Luo H, Marron Fernandez de Velasco E, Wickman K. Neuronal G protein-gated K + channels. Am J Physiol Cell Physiol 2022; 323:C439-C460. [PMID: 35704701 PMCID: PMC9362898 DOI: 10.1152/ajpcell.00102.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels exert a critical inhibitory influence on neurons. Neuronal GIRK channels mediate the G protein-dependent, direct/postsynaptic inhibitory effect of many neurotransmitters including γ-aminobutyric acid (GABA), serotonin, dopamine, adenosine, somatostatin, and enkephalin. In addition to their complex regulation by G proteins, neuronal GIRK channel activity is sensitive to PIP2, phosphorylation, regulator of G protein signaling (RGS) proteins, intracellular Na+ and Ca2+, and cholesterol. The application of genetic and viral manipulations in rodent models, together with recent progress in the development of GIRK channel modulators, has increased our understanding of the physiological and behavioral impact of neuronal GIRK channels. Work in rodent models has also revealed that neuronal GIRK channel activity is modified, transiently or persistently, by various stimuli including exposure drugs of abuse, changes in neuronal activity patterns, and aversive experience. A growing body of preclinical and clinical evidence suggests that dysregulation of GIRK channel activity contributes to neurological diseases and disorders. The primary goals of this review are to highlight fundamental principles of neuronal GIRK channel biology, mechanisms of GIRK channel regulation and plasticity, the nascent landscape of GIRK channel pharmacology, and the potential relevance of GIRK channels to the pathophysiology and treatment of neurological diseases and disorders.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
5
|
Clozapine, nimodipine and endosulfan differentially suppress behavioral defects caused by gain-of-function mutations in a two-pore domain K + channel (UNC-58). Neurosci Res 2020; 170:41-49. [PMID: 32681854 DOI: 10.1016/j.neures.2020.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 11/20/2022]
Abstract
Two-pore domain K+ channels (K2Ps) regulate the resting membrane potential in excitable cells and determine ease of depolarization. Gain-of-function (gf) mutations in one of these channels (unc-58) in C. elegans switch it to a Na+ conductance channel and cause tremors, paralysis and other defects. We hypothesized that it should be possible to identify drugs that corrected these defects in unc-58(gf) mutant animals by blocking or modulating the over-active channels. We examined dispersal of animals on food because the absence of effective forward locomotion is the most obvious defect. In addition, we quantified egg release over 24 h. Starting with a known inhibitor of mammalian K2Ps and directed structure-based screening, we evaluated numerous drugs in these assays. Loratadine, which inhibits human KCNK18, significantly improved movement as did methiothepin. We confirmed that endosulfan, a GABA-A receptor antagonist, corrected locomotion in the unc-58(gf) strains. Based on structural similarities to other hits, we found that clozapine, loxapine and amoxapine potently suppressed abnormal phenotypes. Curiously, nimodipine, a Ca++-channel blocker, dramatically improved movement and egg laying in unc-58(e665), but not unc-58(n495) animals. Molecular modeling provided initial insights into a possible basis for this difference based on the location of the e665 and n495 mutations. This research may lead to identification of novel K2P modulators and potential leads for drug discovery.
Collapse
|
6
|
Serotonin hyperpolarizes the dorsal raphe nucleus neurons of mice by activating G protein–coupled inward rectifier potassium channels. Neuroreport 2020; 31:928-935. [DOI: 10.1097/wnr.0000000000001501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
7
|
Shen WW. Anticraving therapy for alcohol use disorder: A clinical review. Neuropsychopharmacol Rep 2019; 38:105-116. [PMID: 30175522 PMCID: PMC7292332 DOI: 10.1002/npr2.12028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/11/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Aim In this review, the author focused on anticraving therapy for alcohol use disorder (AUD) defined by DMS‐5. A comprehensive review was carried out on the available published papers on anticraving drugs for treating AUD patients. Methods The author described all drugs with anticraving benefits for treating AUD patients approved by the Food and Drug Administration of the United States (US FDA) and European Medicines Agency of the European Union. Then, the commonly prescribed anticraving drugs and those under development were also described. Results The US FDA‐approved anticraving drugs included acamprosate and naltrexone, and those approved by European Medicines Agency were gamma‐hydroxybutyrate and nalmefene. The author also highlighted topiramate, gabapentin, ondansetron, LY196044, ifenprodil, varenicline, ABT‐436, mifepristone, citicoline, and baclofen. The putative mechanisms of action of and the use in clinical practice of those anticraving drugs were also described. Conclusion Although slowly developing, the field of anticraving drugs is getting into shape as a promising entity of a pharmaceutical class of drugs. Then, the author addressed on the underused issues of those recommended, and suggested anticraving drugs by the practice guideline of the American Psychiatric Association. The author urges that clinicians should be more “adventurous” in prescribing those promising drugs because benefits of those anticraving drugs are far‐outweighing the possible side effects of anticraving drugs, or the harms of untreated AUD itself.
Collapse
Affiliation(s)
- Winston W Shen
- Department of Psychiatry, Wan Fang Medical Center, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
8
|
Bérard A, Levin M, Sadler T, Healy D. Selective Serotonin Reuptake Inhibitor Use During Pregnancy and Major Malformations: The Importance of Serotonin for Embryonic Development and the Effect of Serotonin Inhibition on the Occurrence of Malformations. Bioelectricity 2019; 1:18-29. [PMID: 34471805 DOI: 10.1089/bioe.2018.0003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bioelectric signaling is transduced by neurotransmitter pathways in many cell types. One of the key mediators of bioelectric control mechanisms is serotonin, and its transporter SERT, which is targeted by a broad class of blocker drugs (selective serotonin reuptake inhibitors [SSRIs]). Studies showing an increased risk of multiple malformations associated with gestational use of SSRI have been accumulating but debate remains on whether SSRI as a class has the potential to generate these malformations. This review highlights the importance of serotonin for embryonic development; the effect of serotonin inhibition during early pregnancy on the occurrence of multiple diverse malformations that have been shown to occur in human pregnancies; that the risks outweigh the benefits of SSRI use during gestation in populations of mild to moderately depressed pregnant women, which encompass the majority of pregnant depressed women; and that the malformations seen in human pregnancies constitute a pattern of malformations consistent with the known mechanisms of action of SSRIs. We present at least three mechanisms by which SSRI can affect development. These studies highlight the relevance of basic bioelectric and neurotransmitter mechanism for biomedicine.
Collapse
Affiliation(s)
- Anick Bérard
- Faculty of Pharmacy, University of Montreal; Research Center, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology, Medford, Massachusetts
| | - Thomas Sadler
- Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, Utah
| | - David Healy
- Department of Psychiatry, Hergest Unit, Bangor, United Kingdom
| |
Collapse
|
9
|
Constantin S, Wray S. Nociceptin/Orphanin-FQ Inhibits Gonadotropin-Releasing Hormone Neurons via G-Protein-Gated Inwardly Rectifying Potassium Channels. eNeuro 2018; 5:ENEURO.0161-18.2018. [PMID: 30627649 PMCID: PMC6325553 DOI: 10.1523/eneuro.0161-18.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/18/2022] Open
Abstract
The pulsatile release of gonadotropin-releasing hormone (GnRH) is a key feature of the hypothalamic-pituitary-gonadal axis. Kisspeptin neurons in the arcuate nucleus (ARC) trigger GnRH neuronal activity, but how GnRH neurons return to baseline electrical activity is unknown. Nociceptin/orphanin-FQ (OFQ) is an inhibitory neuromodulator. ARC proopiomelanocortin (POMC) neurons, known to receive inputs from ARC kisspeptin neurons, contact GnRH neurons and coexpress OFQ in the rat. In the present study, the effect of OFQ(1-13) on GnRH neurons was determined in the mouse. We identified transcripts for the OFQ receptor [opioid receptor like 1 (ORL1)] in GnRH neurons, and, using two-model systems (explants and slices), we found that OFQ exerted a potent inhibition on GnRH neurons, with or without excitatory inputs. We confirmed that the inhibition was mediated by ORL1 via Gi/o-protein coupling. The inhibition, occurring independently of levels of intracellular cyclic adenosine monophosphate, was sensitive to inwardly rectifying potassium channels. The only specific blocker of Gi/o-protein-coupled inwardly rectifying potassium (GIRK) channels, tertiapin-Q (TPNQ), was ineffective in the inhibition of OFQ. Two GIRK activators, one sharing the binding site of TPNQ and one active only on GIRK1-containing GIRK channels, failed to trigger an inhibition. In contrast, protein kinase C phosphorylation activation, known to inhibit GIRK2-mediated currents, prevented the OFQ inhibition. These results indicate a specific combination of GIRK subunits, GIRK2/3 in GnRH neurons. In vivo, double-labeled OFQ/POMC fibers were found in the vicinity of GnRH neurons, and OFQ fibers apposed GnRH neurons. Together, this study brings to light a potent neuromodulator of GnRH neurons.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, Maryland 20892-3703
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, Maryland 20892-3703
| |
Collapse
|
10
|
Finkelstein Y, Macdonald EM, Li P, Mamdani MM, Gomes T, Juurlink DN. Second-generation anti-depressants and risk of new-onset seizures in the elderly. Clin Toxicol (Phila) 2018; 56:1179-1184. [PMID: 29989445 DOI: 10.1080/15563650.2018.1483025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Anti-depressants are among the most widely-prescribed medications. It is unknown whether the risk of seizure during therapeutic use differs by drug. We ranked the seizure risk of popular anti-depressants. METHODS We conducted a population-based case-control study between April 2002 and March 2015 in Ontario, Canada. Cases were Ontario residents aged ≥65 years hospitalized for a first-ever seizure within 60 d of filling a prescription for one of nine second-generation anti-depressants, each dispensed more than 1 million times (range: 1,196,810 [fluvoxamine] to 19,849,930 [citalopram]) during the study period. For each case, we identified up to four seizure-free controls receiving a similar anti-depressant, and matched on age, sex, date and a pre-defined seizure-specific disease risk index. RESULTS We identified 5701 patients hospitalized with a first-ever seizure and matched them with 21,872 controls. Relative to bupropion, the risk of new-onset seizure during therapeutic use was highest for escitalopram (adjusted odds ratio [OR] 1.79; 95% confidence interval [CI] 1.42-2.25) and citalopram (OR 1.67; 95% CI 1.35-2.07), while no incremental risk was found for fluoxetine (OR 1.02; 95%CI 0.78-1.33) and duloxetine (OR 0.94; 95%CI 0.75-1.22). Other anti-depressants were associated with modest increase in seizure risk. CONCLUSIONS The risk of seizure during therapeutic use among elderly patients varies among second-generation anti-depressants. Escitalopram and citalopram are associated with the highest risk. Prescribers should consider the seizure risk of individual anti-depressants and use discretion when selecting an anti-depressant, especially for patients with other risk factors for seizure. Frontline clinicians should be cognizant of this differential risk.
Collapse
Affiliation(s)
- Yaron Finkelstein
- a Faculty of Medicine, Divisions of Emergency Medicine, Hospital for Sick Children , University of Toronto , Toronto , Ontario , Canada.,b Faculty of Medicine, Department of Clinical Pharmacology and Toxicology, Hospital for Sick Children , University of Toronto , Toronto , Ontario , Canada.,c Child Health Evaluative Sciences , Research Institute, The Hospital for Sick Children , Toronto , Ontario , Canada.,d The Institute for Clinical Evaluative Sciences , Toronto , Ontario , Canada
| | - Erin M Macdonald
- d The Institute for Clinical Evaluative Sciences , Toronto , Ontario , Canada
| | - Ping Li
- d The Institute for Clinical Evaluative Sciences , Toronto , Ontario , Canada
| | - Muhammad M Mamdani
- d The Institute for Clinical Evaluative Sciences , Toronto , Ontario , Canada.,e St. Michael's Hospital , Li Ka Shing Knowledge Institute , Toronto , Ontario , Canada.,f Dalla Lana School of Public Health , University of Toronto , Ontario , Canada.,g Department of Medicine , University of Toronto , Toronto , Ontario , Canada
| | - Tara Gomes
- d The Institute for Clinical Evaluative Sciences , Toronto , Ontario , Canada
| | - David N Juurlink
- d The Institute for Clinical Evaluative Sciences , Toronto , Ontario , Canada.,h Sunnybrook Hospital , Sunnybrook Research Institute , Toronto , Ontario , Canada.,i Departments of Medicine, Pediatrics and Health Policy, Management and Evaluation , University of Toronto , Ontario , Canada
| |
Collapse
|
11
|
Lack of relationship between plasma levels of escitalopram and QTc-interval length. Eur Arch Psychiatry Clin Neurosci 2017; 267:815-822. [PMID: 28116499 DOI: 10.1007/s00406-016-0758-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 12/13/2016] [Indexed: 10/20/2022]
Abstract
Despite safety concerns raised by the European Medicines Agency (EMA), evidence supporting QT-lengthening effects of escitalopram is far to be conclusive. We aimed to evaluate the relationship between escitalopram plasma levels (Escit-PL) and corrected QT-interval length (QTc-length) in 91 outpatients recruited from a hospital setting. Fifteen patients had an abnormally prolonged QTc-interval, and 3 had QTc-intervals ≥500 ms. No correlation between Escit-PL and QTc-length was found (r = 0.08; p = 0.45). Linear/logistic regression analyses were also conducted taking into account potential confounders such as age, gender, personal history of heart disease, medication load and concomitant use of antipsychotic/tricyclic antidepressants. Escit-PL did not predict either QTc-length or abnormally prolonged QTc-interval. Only antipsychotics/tricyclics use (adjusted β = 0.26, SE = 9.1; p = 0.01) was an independent predictor of QTc-length (R 2 = 0.096, F = 4.68, df = 2,88; p = 0.01). Only antipsychotics/tricyclics use (OR 3.56 [95% CI 1.01-12.52]; p < 0.05) and medication load (OR 1.32 [95% CI 1.06-1.64]; p < 0.01) were significantly associated with an increased risk of abnormally prolonged QTc-interval (Omnibus test χ 2 = 9.5, df = 2; p < 0.01). Our study did not find a significant relationship between Escit-PL and QTc-length even when recognized modulating factors of the QT-interval were controlled for. Concomitant use of other potentially arrhythmogenic agents may help to explain the apparent link between escitalopram and QT prolongation previously suggested. The advisability of maintaining the EMA warning is once again called into question.
Collapse
|
12
|
Frizzo ME. Can a Selective Serotonin Reuptake Inhibitor Act as a Glutamatergic Modulator? Curr Ther Res Clin Exp 2017; 87:9-12. [PMID: 28912901 PMCID: PMC5583143 DOI: 10.1016/j.curtheres.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2017] [Indexed: 01/05/2023] Open
Abstract
Sertraline (Zoloft) and fluoxetine (Prozac) are selective serotonin reuptake inhibitors whose antidepressant mechanism of action is classically attributed to an elevation of the extracellular levels of serotonin in the synaptic cleft. However, the biological effects of these drugs seem to be more complex than their traditionally described mechanism of action. Among their actions is the inhibition of different types of Na+ and K+ channels, as well as of glutamate uptake activity. The clearance of extracellular glutamate is essential to maintain the central nervous system within physiological conditions, and this excitatory neurotransmitter is removed from the synaptic cleft by astrocyte transporters. This transport depends upon a hyperpolarized membrane potential in astrocytes that is mainly maintained by Kir4.1 K+ channels. The impairment of the Kir4.1 channel activity reduces driving force for the glutamate transporter, resulting in an accumulation of extracellular glutamate. It has been shown that sertraline and fluoxetine inhibit Kir4.1 K+ channels. Recently, we demonstrated that sertraline reduces glutamate uptake in human platelets, which contain a high-affinity Na+-dependent glutamate uptake system, with kinetic and pharmacological properties similar to astrocytes in the central nervous system. Considering these similarities between human platelets and astrocytes, one might ask if sertraline could potentially reduce glutamate clearance in the synaptic cleft and consequently modulate glutamatergic transmission. This possibility merits investigation, since it may provide additional information regarding the mechanism of action and perhaps the side effects of these antidepressants.
Collapse
Affiliation(s)
- Marcos Emilio Frizzo
- Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500. CEP 90050-170, Porto Alegre, Brazil
| |
Collapse
|
13
|
Dual activation of neuronal G protein-gated inwardly rectifying potassium (GIRK) channels by cholesterol and alcohol. Sci Rep 2017; 7:4592. [PMID: 28676630 PMCID: PMC5496853 DOI: 10.1038/s41598-017-04681-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/18/2017] [Indexed: 12/20/2022] Open
Abstract
Activation of G protein-gated inwardly rectifying potassium (GIRK) channels leads to a hyperpolarization of the neuron’s membrane potential, providing an important component of inhibition in the brain. In addition to the canonical G protein-activation pathway, GIRK channels are activated by small molecules but less is known about the underlying gating mechanisms. One drawback to previous studies has been the inability to control intrinsic and extrinsic factors. Here we used a reconstitution strategy with highly purified mammalian GIRK2 channels incorporated into liposomes and demonstrate that cholesterol or intoxicating concentrations of ethanol, i.e., >20 mM, each activate GIRK2 channels directly, in the absence of G proteins. Notably, both activators require the membrane phospholipid PIP2 but appear to interact independently with different regions of the channel. Elucidating the mechanisms underlying G protein-independent pathways of activating GIRK channels provides a unique strategy for developing new types of neuronal excitability modulators.
Collapse
|
14
|
Abstract
Toxicity attributed to sertraline has been demonstrated recently in different cell types and also in some organisms. We investigated the effect of sertraline on planarians, which are considered suitable for investigations in neurotoxicology and currently are widely used as an animal model in neuropharmacological studies. Planarians treated with 10 µM sertraline showed a rapid reduction in their spontaneous movement until they became completely motionless and then showed a series of asynchronous paroxysms (seizures) followed by progressive tissue damage, beginning 48 h after the sertraline treatment, and died approximately 72 h later. Our data showed that sertraline does not cause planarian death within the range of therapeutic concentrations; however, behavioral alterations were observed with concentrations that can be considered compatible with therapeutic ones, such as a significant reduction in planarian locomotory activity at 0.4 µM. Treatment with 4 µM sertraline had a significant effect, reducing planarian locomotory activity and increasing the number of asynchronous paroxysms; both effects were significantly maintained even 24 h after the sertraline was withdrawn. These behavioral changes observed at low micromolar concentrations suggest that sertraline might have residual biological consequences for planarians, even after it is withdrawn.
Collapse
|
15
|
Kim HS, Li H, Kim HW, Shin SE, Choi IW, Firth AL, Bang H, Bae YM, Park WS. Selective serotonin reuptake inhibitor sertraline inhibits voltage-dependent K+ channels in rabbit coronary arterial smooth muscle cells. J Biosci 2017; 41:659-666. [PMID: 27966486 DOI: 10.1007/s12038-016-9645-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We examined the effects of the selective serotonin reuptake inhibitor (SSRI) sertraline on voltage-dependent K+ (Kv) channels in freshly isolated rabbit coronary arterial smooth muscle cells using the voltage-clamp technique. Sertraline decreased the Kv channel current in a dose-dependent manner, with an IC50 value of 0.18 mu M and a slope value (Hill coefficient) of 0.61. Although the application of 1 mu M sertraline did not affect the steady-state activation curves, sertraline caused a significant, negative shift in the inactivation curves. Pretreatment with another SSRI, paroxetine, had no significant effect on Kv currents and did not alter the inhibitory effects of sertraline on Kv currents. From these results, we concluded that sertraline dose-dependently inhibited Kv currents independently of serotonin reuptake inhibition by shifting inactivation curves to a more negative potential.
Collapse
Affiliation(s)
- Han Sol Kim
- Department of Physiology, Kangwon National University School of Medicine Chuncheon 200-701, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mitragynine and its potential blocking effects on specific cardiac potassium channels. Toxicol Appl Pharmacol 2016; 305:22-39. [PMID: 27260674 DOI: 10.1016/j.taap.2016.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 02/07/2023]
Abstract
Mitragyna speciosa Korth is known for its euphoric properties and is frequently used for recreational purposes. Several poisoning and fatal cases involving mitragynine have been reported but the underlying causes remain unclear. Human ether-a-go-go-related gene (hERG) encodes the cardiac IKr current which is a determinant of the duration of ventricular action potentials and QT interval. On the other hand, IK1, a Kir current mediated by Kir2.1 channel and IKACh, a receptor-activated Kir current mediated by GIRK channel are also known to be important in maintaining the cardiac function. This study investigated the effects of mitragynine on the current, mRNA and protein expression of hERG channel in hERG-transfected HEK293 cells and Xenopus oocytes. The effects on Kir2.1 and GIRK channels currents were also determined in the oocytes. The hERG tail currents following depolarization pulses were inhibited by mitragynine with an IC50 value of 1.62μM and 1.15μM in the transfected cell line and Xenopus oocytes, respectively. The S6 point mutations of Y652A and F656A attenuated the inhibitor effects of mitragynine, indicating that mitragynine interacts with these high affinity drug-binding sites in the hERG channel pore cavity which was consistent with the molecular docking simulation. Interestingly, mitragynine does not affect the hERG expression at the transcriptional level but inhibits the protein expression. Mitragynine is also found to inhibit IKACh current with an IC50 value of 3.32μM but has no significant effects on IK1. Blocking of both hERG and GIRK channels may cause additive cardiotoxicity risks.
Collapse
|
17
|
Glaaser IW, Slesinger PA. Structural Insights into GIRK Channel Function. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:117-60. [PMID: 26422984 DOI: 10.1016/bs.irn.2015.05.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
G protein-gated inwardly rectifying potassium (GIRK; Kir3) channels, which are members of the large family of inwardly rectifying potassium channels (Kir1-Kir7), regulate excitability in the heart and brain. GIRK channels are activated following stimulation of G protein-coupled receptors that couple to the G(i/o) (pertussis toxin-sensitive) G proteins. GIRK channels, like all other Kir channels, possess an extrinsic mechanism of inward rectification involving intracellular Mg(2+) and polyamines that occlude the conduction pathway at membrane potentials positive to E(K). In the past 17 years, more than 20 high-resolution atomic structures containing GIRK channel cytoplasmic domains and transmembrane domains have been solved. These structures have provided valuable insights into the structural determinants of many of the properties common to all inward rectifiers, such as permeation and rectification, as well as revealing the structural bases for GIRK channel gating. In this chapter, we describe advances in our understanding of GIRK channel function based on recent high-resolution atomic structures of inwardly rectifying K(+) channels discussed in the context of classical structure-function experiments.
Collapse
Affiliation(s)
- Ian W Glaaser
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul A Slesinger
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
18
|
Balaraman Y, Lahiri DK, Nurnberger JI. Variants in Ion Channel Genes Link Phenotypic Features of Bipolar Illness to Specific Neurobiological Process Domains. MOLECULAR NEUROPSYCHIATRY 2015; 1:23-35. [PMID: 27602355 DOI: 10.1159/000371886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022]
Abstract
Recent advances in genome-wide association studies are pointing towards a major role for voltage-gated ion channels in neuropsychiatric disorders and, in particular, bipolar disorder (BD). The phenotype of BD is complex, with symptoms during mood episodes and deficits persisting between episodes. We have tried to elucidate the common neurobiological mechanisms associated with ion channel signaling in order to provide a new perspective on the clinical symptoms and possible endophenotypes seen in BD patients. We propose a model in which the multiple variants in genes coding for ion channel proteins would perturb motivational circuits, synaptic plasticity, myelination, hypothalamic-pituitary-adrenal axis function, circadian neuronal rhythms, and energy regulation. These changes in neurobiological mechanisms would manifest in endophenotypes of aberrant reward processing, white matter hyperintensities, deficits in executive function, altered frontolimbic connectivity, increased amygdala activity, increased melatonin suppression, decreased REM latency, and aberrant myo-inositol/ATP shuttling. The endophenotypes result in behaviors of poor impulse control, motivational changes, cognitive deficits, abnormal stress response, sleep disturbances, and energy changes involving different neurobiological process domains. The hypothesis is that these disturbances start with altered neural circuitry during development, following which multiple environmental triggers may disrupt the neuronal excitability balance through an activity-dependent molecular process, resulting in clinical mood episodes.
Collapse
Affiliation(s)
- Yokesh Balaraman
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - Debomoy K Lahiri
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - John I Nurnberger
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| |
Collapse
|
19
|
Bodhinathan K, Slesinger PA. Alcohol modulation of G-protein-gated inwardly rectifying potassium channels: from binding to therapeutics. Front Physiol 2014; 5:76. [PMID: 24611054 PMCID: PMC3933770 DOI: 10.3389/fphys.2014.00076] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/07/2014] [Indexed: 12/27/2022] Open
Abstract
Alcohol (ethanol)-induced behaviors may arise from direct interaction of alcohol with discrete protein cavities within brain proteins. Recent structural and biochemical studies have provided new insights into the mechanism of alcohol-dependent activation of G protein-gated inwardly rectifying potassium (GIRK) channels, which regulate neuronal responses in the brain reward circuit. GIRK channels contain an alcohol binding pocket formed at the interface of two adjacent channel subunits. Here, we discuss the physiochemical properties of the alcohol pocket and the roles of G protein βγ subunits and membrane phospholipid PIP2 in regulating the alcohol response of GIRK channels. Some of the features of alcohol modulation of GIRK channels may be common to other alcohol-sensitive brain proteins. We discuss the possibility of alcohol-selective therapeutics that block alcohol access to the pocket. Understanding alcohol recognition and modulation of brain proteins is essential for development of therapeutics for alcohol abuse and addiction.
Collapse
Affiliation(s)
- Karthik Bodhinathan
- Structural Biology and Peptide Biology Laboratories, The Salk Institute for Biological Studies La Jolla, CA, USA
| | - Paul A Slesinger
- Structural Biology and Peptide Biology Laboratories, The Salk Institute for Biological Studies La Jolla, CA, USA ; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York, NY, USA
| |
Collapse
|
20
|
[Citalopram, escitalopram and prolonged QT: warning or alarm?]. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2014; 7:147-50. [PMID: 24556340 DOI: 10.1016/j.rpsm.2013.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 11/25/2013] [Accepted: 12/24/2013] [Indexed: 11/20/2022]
Abstract
The alerts issued by regulatory agencies on the potential cardiac toxicity of citalopram and escitalopram have caused alarm among clinicians. A review of the data concerning this topic shows that the alarm should be limited to patients with a history of syncope or poisoning. As a precautionary measure, an electrocardiogram should be performed on elderly patients.
Collapse
|
21
|
Hsu YC, Tzeng NS. Can topical duloxetine be used for neuropathic pain? Med Hypotheses 2014; 82:175-6. [DOI: 10.1016/j.mehy.2013.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/04/2013] [Accepted: 11/19/2013] [Indexed: 11/25/2022]
|
22
|
Reeves KC, Virk S, Niedermier J, Duchemin AM. Addition of amoxapine improves positive and negative symptoms in a patient with schizophrenia. Ther Adv Psychopharmacol 2013; 3:340-2. [PMID: 24294487 PMCID: PMC3840811 DOI: 10.1177/2045125313499363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kevin C Reeves
- Wexner Medical at the Ohio State University, 1670 Upham Drive, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
23
|
Luján R, Marron Fernandez de Velasco E, Aguado C, Wickman K. New insights into the therapeutic potential of Girk channels. Trends Neurosci 2013; 37:20-9. [PMID: 24268819 DOI: 10.1016/j.tins.2013.10.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 01/01/2023]
Abstract
G protein-dependent signaling pathways control the activity of excitable cells of the nervous system and heart, and are the targets of neurotransmitters, clinically relevant drugs, and drugs of abuse. G protein-gated inwardly rectifying potassium (K(+)) (Girk/Kir3) channels are a key effector in inhibitory signaling pathways. Girk-dependent signaling contributes to nociception and analgesia, reward-related behavior, mood, cognition, and heart-rate regulation, and has been linked to epilepsy, Down syndrome, addiction, and arrhythmias. We discuss recent advances in our understanding of Girk channel structure, organization in signaling complexes, and plasticity, as well as progress on the development of subunit-selective Girk modulators. These findings offer new hope for the selective manipulation of Girk channels to treat a variety of debilitating afflictions.
Collapse
Affiliation(s)
- Rafael Luján
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain.
| | | | - Carolina Aguado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, 321 Church Street South East, Minneapolis, MN 55455, USA.
| |
Collapse
|
24
|
Fischer F, Vonderlin N, Seyler C, Zitron E, Schernus B, Katus H, Scholz E. Acute and subacute effects of the selective serotonin–noradrenaline reuptake inhibitor duloxetine on cardiac hERG channels. Naunyn Schmiedebergs Arch Pharmacol 2013; 386:795-804. [DOI: 10.1007/s00210-013-0878-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/22/2013] [Indexed: 11/28/2022]
|
25
|
Scuteri A, Modestino A, Fedullo F, Assisi AP, Gianni W. Depression treatment selectively modifies arterial stiffness in older participants. J Gerontol A Biol Sci Med Sci 2012; 68:719-25. [PMID: 23160364 DOI: 10.1093/gerona/gls230] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Depression is emerging as an independent cardiovascular disease risk factor. We investigated whether treating depression in older participants impacted on arterial stiffness, a known cardiovascular disease risk factor and a clinical marker of arterial aging. METHODS Seventy-five participants with pulse wave velocity (PWV), the gold standard measure for arterial stiffness, at baseline and at 12-month follow-up were included. Depressed patients were randomized to escitalopram (10mg/d) or to duloxetine (60mg/d). In patients without depression, no antidepressant therapy was started. The psychologist and the doctor measuring PWV were both unaware of antidepressant treatment. RESULTS At study entry, no difference in PWV were observable in the three groups of participants. A significant time × drug interaction term (p < .05) was observed for the impact of antidepressant therapy on PWV by analysis of covariance analysis. After 12 months of therapy, duloxetine treatment resulted in a significant (+21%) and escitalopram treatment in a not significant (6%) PWV increase. These changes in PWV were accompanied by a similar increase in blood pressure and LDL cholesterol in the two treated groups. However, duloxetine resulted in a significant 10% greater heart rate after 12 months that was not observable in participants treated with escitalopram nor in not-depressed older participants. Multiple regression models revealed that a drug-specific effect on PWV persisted after controlling for cardiovascular risk factor levels. CONCLUSION Duloxetine but not escitalopram significantly increased PWV in older depressed participants after 12 months of treatment. The effect was not fully explained by concomitant changes in traditional cardiovascular risk factors known to significantly impact arterial stiffness.
Collapse
|
26
|
Lee HA, Kim KS, Hyun SA, Park SG, Kim SJ. Wide spectrum of inhibitory effects of sertraline on cardiac ion channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:327-32. [PMID: 23118556 PMCID: PMC3484517 DOI: 10.4196/kjpp.2012.16.5.327] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 09/11/2012] [Accepted: 09/11/2012] [Indexed: 01/21/2023]
Abstract
Sertraline is a commonly used antidepressant of the selective serotonin reuptake inhibitors (SSRIs) class. In these experiments, we have used the whole cell patch clamp technique to examine the effects of sertraline on the major cardiac ion channels expressed in HEK293 cells and the native voltage-gated Ca(2+) channels in rat ventricular myocytes. According to the results, sertraline is a potent blocker of cardiac K(+) channels, such as hERG, I(Ks) and I(K1). The rank order of inhibitory potency was hERG >I(K1)> I(Ks) with IC(50) values of 0.7, 10.5, and 15.2 µM, respectively. In addition to K(+) channels, sertraline also inhibited I(Na) and I(Ca), and the IC(50) values are 6.1 and 2.6 µM, respectively. Modification of these ion channels by sertraline could induce changes of the cardiac action potential duration and QT interval, and might result in cardiac arrhythmia.
Collapse
Affiliation(s)
- Hyang-Ae Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejeon 305-600, Korea. ; Department of Physiology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | | | | | |
Collapse
|
27
|
Wang CL, Tsai ML, Wu SN. Evidence for mitoxantrone-induced block of inwardly rectifying K(+) channels expressed in the osteoclast precursor RAW 264.7 cells differentiated with lipopolysaccharide. Cell Physiol Biochem 2012; 30:687-701. [PMID: 22854649 DOI: 10.1159/000341449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Mitoxanthrone (MX) is an anthracenedione antineoplastic agent. Whether this drug and other related compounds have any effects on ion currents in osteoclasts remains largely unclear. METHODS In this study, the effects of MX and other related compounds on inwardly rectifying K(+) current (I(K(IR))) were investigated in RAW 264.7 osteoclast precursor cells treated with lipopolysaccharide. RESULTS The I(K(IR))in these cells are blocked by BaCl(2) (1 mM). MX (1-100 µM) decreased the amplitude of I(K(IR)) in a concentration-dependent manner with an IC(50) value of 6.4 µM. MX also slowed the time course of I(K(IR)) inactivation elicited by large hyperpolarization. Doxorubicin (10 µM), 17β-estradiol (10 µM) and tertiapin (1 µM) decreased the I(K(IR)) amplitude in these cells. In bafilomycin A(1)-treated cells, MX-mediated block of I(K(IR)) still existed. In cell-attached configuration, when the electrode was filled with MX (10 µM), the activity of inwardly rectifying K(+) (Kir) channels was decreased with no change in single-channel conductance. MX-mediated reduction of channel activity is accompanied by a shortening of mean open time. Under current-clamp conditions, addition of MX resulted in membrane depolarization. Therefore, MX can interact with the Kir channels to decrease the I(K(IR)) amplitude and to depolarize the membrane in these cells. CONCLUSION The block by this drug of Kir2.1 channels appears to be one of the important mechanisms underlying its actions on the resorptive activity of osteoclasts, if similar results occur in vivo. Targeting at Kir channels may be clinically useful as an adjunctive regimen to anti-cancer drugs (e.g., MX or doxorubicin) in influencing the resorptive activity of osteoclasts.
Collapse
Affiliation(s)
- Chung-Lin Wang
- Department of Nursing, Chung Jen College of Nursing, Health Sciences and Management, Chiayi city, Taiwan
| | | | | |
Collapse
|