1
|
Gazulla J, Berciano J. Potential Benefit of Channel Activators in Loss-of-Function Primary Potassium Channelopathies Causing Heredoataxia. CEREBELLUM (LONDON, ENGLAND) 2024; 23:833-837. [PMID: 37460907 DOI: 10.1007/s12311-023-01584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 03/20/2024]
Abstract
Potassium channels (KCN) are transmembrane complexes that regulate the resting membrane potential and the duration of action potentials in cells. The opening of KCN brings about an efflux of K+ ions that induces cell repolarization after depolarization, returns the transmembrane potential to its resting state, and enables for continuous spiking ability. The aim of this work was to assess the role of KCN dysfunction in the pathogenesis of hereditary ataxias and the mechanisms of action of KCN opening agents (KCO). In consequence, a review of the ad hoc medical literature was performed. Among hereditary KCN diseases causing ataxia, mutated Kv3.3, Kv4.3, and Kv1.1 channels provoke spinocerebellar ataxia (SCA) type 13, SCA19/22, and episodic ataxia type 1 (EA1), respectively. The K+ efflux was found to be reduced in experimental models of these diseases, resulting in abnormally prolonged depolarization and incomplete repolarization, thereby interfering with repetitive discharges in the cells. Hence, substances able to promote normal spiking activity in the cerebellum could provide symptomatic benefit. Although drugs used in clinical practice do not activate Kv3.3 or Kv4.3 directly, available KCO probably could ameliorate ataxic symptoms in SCA13 and SCA19/22, as verified with acetazolamide in EA1, and retigabine in a mouse model of hypokalemic periodic paralysis. To summarize, ataxia could possibly be improved by non-specific KCO in SCA13 and SCA19/22. The identification of new specific KCO agents will undoubtedly constitute a promising therapeutic strategy for these diseases.
Collapse
Affiliation(s)
- José Gazulla
- Department of Neurology, Hospital Universitario Miguel Servet, Isabel la Católica, 1-3, 50009, Saragossa, Spain.
| | - José Berciano
- Department of Neurology, Hospital Universitario Marqués de Valdecilla (IDIVAL), University of Cantabria, CIBERNED, Avenida de Valdecilla S/N, 39008, Santander, Spain
| |
Collapse
|
2
|
Kong DW, Du LD, Liu RZ, Yuan TY, Wang SB, Wang YH, Lu Y, Fang LH, Du GH. Baicalein attenuates rotenone-induced SH-SY5Y cell apoptosis through binding to SUR1 and activating ATP-sensitive potassium channels. Acta Pharmacol Sin 2024; 45:480-489. [PMID: 37993535 PMCID: PMC10834402 DOI: 10.1038/s41401-023-01187-3] [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: 05/04/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023] Open
Abstract
Dopaminergic neurons in the substantia nigra (SN) expressing SUR1/Kir6.2 type ATP-sensitive potassium channels (K-ATP) are more vulnerable to rotenone or metabolic stress, which may be an important reason for the selective degeneration of neurons in Parkinson's disease (PD). Baicalein has shown neuroprotective effects in PD animal models. In this study, we investigated the effect of baicalein on K-ATP channels and the underlying mechanisms in rotenone-induced apoptosis of SH-SY5Y cells. K-ATP currents were recorded from SH-SY5Y cells using whole-cell voltage-clamp recording. Drugs dissolved in the external solution at the final concentration were directly pipetted onto the cells. We showed that rotenone and baicalein opened K-ATP channels and increased the current amplitudes with EC50 values of 0.438 μM and 6.159 μM, respectively. K-ATP channel blockers glibenclamide (50 μM) or 5-hydroxydecanoate (5-HD, 250 μM) attenuated the protective effects of baicalein in reducing reactive oxygen species (ROS) content and increasing mitochondrial membrane potential and ATP levels in rotenone-injured SH-SY5Y cells, suggesting that baicalein protected against the apoptosis of SH-SY5Y cells by regulating the effect of rotenone on opening K-ATP channels. Administration of baicalein (150, 300 mg·kg-1·d-1, i.g.) significantly inhibited rotenone-induced overexpression of SUR1 in SN and striatum of rats. We conducted surface plasmon resonance assay and molecular docking, and found that baicalein had a higher affinity with SUR1 protein (KD = 10.39 μM) than glibenclamide (KD = 24.32 μM), thus reducing the sensitivity of K-ATP channels to rotenone. Knockdown of SUR1 subunit reduced rotenone-induced apoptosis and damage of SH-SY5Y cells, confirming that SUR1 was an important target for slowing dopaminergic neuronal degeneration in PD. Taken together, we demonstrate for the first time that baicalein attenuates rotenone-induced SH-SY5Y cell apoptosis through binding to SUR1 and activating K-ATP channels.
Collapse
Affiliation(s)
- De-Wen Kong
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Li-da Du
- Shandong Soteria Pharmaceutical Co Ltd., Jinan, 250022, China
| | - Run-Zhe Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tian-Yi Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shou-Bao Wang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yue-Hua Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yang Lu
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lian-Hua Fang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
3
|
Lai H, Gao M, Yang H. The potassium channels: Neurobiology and pharmacology of tinnitus. J Neurosci Res 2024; 102:e25281. [PMID: 38284861 DOI: 10.1002/jnr.25281] [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: 04/23/2023] [Revised: 10/27/2023] [Accepted: 11/16/2023] [Indexed: 01/30/2024]
Abstract
Tinnitus is a widespread public health issue that imposes a significant social burden. The occurrence and maintenance of tinnitus have been shown to be associated with abnormal neuronal activity in the auditory pathway. Based on this view, neurobiological and pharmacological developments in tinnitus focus on ion channels and synaptic neurotransmitter receptors in neurons in the auditory pathway. With major breakthroughs in the pathophysiology and research methodology of tinnitus in recent years, the role of the largest family of ion channels, potassium ion channels, in modulating the excitability of neurons involved in tinnitus has been increasingly demonstrated. More and more potassium channels involved in the neural mechanism of tinnitus have been discovered, and corresponding drugs have been developed. In this article, we review animal (mouse, rat, hamster, and guinea-pig), human, and genetic studies on the different potassium channels involved in tinnitus, analyze the limitations of current clinical research on potassium channels, and propose future prospects. The aim of this review is to promote the understanding of the role of potassium ion channels in tinnitus and to advance the development of drugs targeting potassium ion channels for tinnitus.
Collapse
Affiliation(s)
- Haohong Lai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minqian Gao
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Hearing and Speech-Language Science, Guangzhou Xinhua University, Guangzhou, China
| | - Haidi Yang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Hearing and Speech-Language Science, Guangzhou Xinhua University, Guangzhou, China
| |
Collapse
|
4
|
Kwon YJ, Kwon OI, Hwang HJ, Shin HC, Yang S. Therapeutic effects of phlorotannins in the treatment of neurodegenerative disorders. Front Mol Neurosci 2023; 16:1193590. [PMID: 37305552 PMCID: PMC10249478 DOI: 10.3389/fnmol.2023.1193590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Phlorotannins are natural polyphenolic compounds produced by brown marine algae and are currently found in nutritional supplements. Although they are known to cross the blood-brain barrier, their neuropharmacological actions remain unclear. Here we review the potential therapeutic benefits of phlorotannins in the treatment of neurodegenerative diseases. In mouse models of Alzheimer's disease, ethanol intoxication and fear stress, the phlorotannin monomer phloroglucinol and the compounds eckol, dieckol and phlorofucofuroeckol A have been shown to improve cognitive function. In a mouse model of Parkinson's disease, phloroglucinol treatment led to improved motor performance. Additional neurological benefits associated with phlorotannin intake have been demonstrated in stroke, sleep disorders, and pain response. These effects may stem from the inhibition of disease-inducing plaque synthesis and aggregation, suppression of microglial activation, modulation of pro-inflammatory signaling, reduction of glutamate-induced excitotoxicity, and scavenging of reactive oxygen species. Clinical trials of phlorotannins have not reported significant adverse effects, suggesting these compounds to be promising bioactive agents in the treatment of neurological diseases. We therefore propose a putative biophysical mechanism of phlorotannin action in addition to future directions for phlorotannin research.
Collapse
Affiliation(s)
- Yoon Ji Kwon
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Oh Ig Kwon
- Botamedi Brain Health and Medical Care Company Limited, Central, Hong Kong SAR, China
| | - Hye Jeong Hwang
- Center for Molecular Intelligence, SUNY Korea, Incheon, Republic of Korea
| | - Hyeon-Cheol Shin
- Botamedi Brain Health and Medical Care Company Limited, Central, Hong Kong SAR, China
- Center for Molecular Intelligence, SUNY Korea, Incheon, Republic of Korea
| | - Sungchil Yang
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| |
Collapse
|
5
|
Noh W, Pak S, Choi G, Yang S, Yang S. Transient Potassium Channels: Therapeutic Targets for Brain Disorders. Front Cell Neurosci 2019; 13:265. [PMID: 31263403 PMCID: PMC6585177 DOI: 10.3389/fncel.2019.00265] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/28/2019] [Indexed: 01/04/2023] Open
Abstract
Transient potassium current channels (IA channels), which are expressed in most brain areas, have a central role in modulating feedforward and feedback inhibition along the dendroaxonic axis. Loss of the modulatory channels is tightly associated with a number of brain diseases such as Alzheimer’s disease, epilepsy, fragile X syndrome (FXS), Parkinson’s disease, chronic pain, tinnitus, and ataxia. However, the functional significance of IA channels in these diseases has so far been underestimated. In this review, we discuss the distribution and function of IA channels. Particularly, we posit that downregulation of IA channels results in neuronal (mostly dendritic) hyperexcitability accompanied by the imbalanced excitation and inhibition ratio in the brain’s networks, eventually causing the brain diseases. Finally, we propose a potential therapeutic target: the enhanced action of IA channels to counteract Ca2+-permeable channels including NMDA receptors could be harnessed to restore dendritic excitability, leading to a balanced neuronal state.
Collapse
Affiliation(s)
- Wonjun Noh
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| | - Sojeong Pak
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Geunho Choi
- Department of Computer Science and Engineering, Incheon National University, Incheon, South Korea
| | - Sungchil Yang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Sunggu Yang
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| |
Collapse
|
6
|
Lin Z, Huang X, Zhou W, Zhang W, Liu Y, Bian T, Niu L, Meng L, Guo Y. Ultrasound Stimulation Modulates Voltage-Gated Potassium Currents Associated With Action Potential Shape in Hippocampal CA1 Pyramidal Neurons. Front Pharmacol 2019; 10:544. [PMID: 31178727 PMCID: PMC6538798 DOI: 10.3389/fphar.2019.00544] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/30/2019] [Indexed: 11/28/2022] Open
Abstract
Potassium channels (K+) play an important role in the regulation of cellular signaling. Dysfunction of potassium channels is associated with several severe ion channels diseases, such as long QT syndrome, episodic ataxia and epilepsy. Ultrasound stimulation has proven to be an effective non-invasive tool for the modulation of ion channels and neural activity. In this study, we demonstrate that ultrasound stimulation enables to modulate the potassium currents and has an impact on the shape modulation of action potentials (AP) in the hippocampal pyramidal neurons using whole-cell patch-clamp recordings in vitro. The results show that outward potassium currents in neurons increase significantly, approximately 13%, in response to 30 s ultrasound stimulation. Simultaneously, the increasing outward potassium currents directly decrease the resting membrane potential (RMP) from −64.67 ± 1.10 mV to −67.51 ± 1.35 mV. Moreover, the threshold current and AP fall rate increase while the reduction of AP half-width and after-hyperpolarization peak time is detected. During ultrasound stimulation, reduction of the membrane input resistance of pyramidal neurons can be found and shorter membrane time constant is achieved. Additionally, we verify that the regulation of potassium currents and shape of action potential is mainly due to the mechanical effects induced by ultrasound. Therefore, ultrasound stimulation may offer an alternative tool to treat some ion channels diseases related to potassium channels.
Collapse
Affiliation(s)
- Zhengrong Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaowei Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wenjun Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of E&M, Ministry of Education and Zhejiang Province, Zhejiang University of Technology, Hangzhou, China
| | - Yingzhe Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Tianyuan Bian
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Sino-Dutch Biomedical and Information Engineering School, Northeastern University, Shenyang, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering - Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 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, China
| |
Collapse
|
7
|
Judy JT, Zandi PP. A review of potassium channels in bipolar disorder. Front Genet 2013; 4:105. [PMID: 23781230 PMCID: PMC3678088 DOI: 10.3389/fgene.2013.00105] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/22/2013] [Indexed: 12/11/2022] Open
Abstract
Although bipolar disorder (BP) is one of the most heritable psychiatric conditions, susceptibility genes for the disorder have yet to be conclusively identified. It is likely that variants in multiple genes across multiple pathways contribute to the genotype–phenotype relationship in the affected population. Recent evidence from genome-wide association studies implicates an entire class of genes related to the structure and regulation of ion channels, suggesting that the etiology of BP may arise from channelopathies. In this review, we examine the evidence for this hypothesis, with a focus on the potential role of voltage-gated potassium channels. We consider evidence from genetic and expression studies, and discuss the potential underlying biology. We consider animal models and treatment implications of the involvement of potassium ion channelopathy in BP. Finally, we explore intriguing parallels between BP and epilepsy, the signature channelopathy of the central nervous system.
Collapse
Affiliation(s)
- Jennifer T Judy
- Department of Psychiatry, Johns Hopkins School of Medicine Baltimore, MD, USA
| | | |
Collapse
|
8
|
Lee BC, Lim HH, Kim S, Youn HS, Lee Y, Kim YC, Eom SH, Lee KW, Park CS. Localization of a site of action for benzofuroindole-induced potentiation of BKCa channels. Mol Pharmacol 2012; 82:143-55. [PMID: 22547262 DOI: 10.1124/mol.112.078097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
As previously reported, the activity of the large-conductance calcium (Ca(2+))-activated potassium (K(+)) (BK(Ca)) channel is strongly potentiated from the extracellular side of the cell membrane by certain benzofuroindole derivatives. Here, the mechanism of action of one of the most potent activators, 4-chloro-7-(trifluoromethyl)-10H-benzofuro[3,2-b]indole-1-carboxylic acid (CTBIC), is characterized. This compound, Compound 22 in the previous report (Chembiochem 6:1745-1748, 2005), potentiated the activity of the channel by shifting its conductance-voltage relationship toward the more negative direction. Cotreatment with CTBIC reduced the affinity of charybdotoxin, a peptide pore-blocker, whereas that of tetraethylammonium, a small pore-blocking quaternary ammonium, was not significantly altered. Guided by these results, scanning mutagenesis of the outer vestibule of the BK(Ca) channel was launched to uncover the molecular determinants that affect CTBIC binding. Alanine substitution of several amino acid residues in the turret region and the S6 helix of the channel decreased potentiation by CTBIC. Homology modeling and molecular dynamics simulation showed that some of these residues formed a CTBIC binding pocket between two adjacent α-subunits in the outer vestibule of the channel. Thus, it can be envisioned that benzofuroindole derivatives stabilize the open conformation of the channel by binding to the residues clustered across the extracellular part of the subunit interface. The present results indicate that the interface between different α-subunits of the BK(Ca) channel may play a critical role in the modulation of channel activity. Therefore, this interface represents a potential therapeutic target site for the regulation of K(+) channels.
Collapse
Affiliation(s)
- Byoung-Cheol Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), 123 Cheomdangwagi-ro, Buk-gu, Gwangju, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Choi SH, Shin TJ, Lee BH, Hwang SH, Lee SM, Lee BC, Park CS, Ha TS, Nah SY. Ginsenoside Rg3 enhances large conductance Ca2+-activated potassium channel currents: a role of Tyr360 residue. Mol Cells 2011; 31:133-40. [PMID: 21191818 PMCID: PMC3932677 DOI: 10.1007/s10059-011-0017-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 10/25/2010] [Accepted: 11/13/2010] [Indexed: 10/18/2022] Open
Abstract
Ginsenosides, active ingredients of Panax ginseng, are known to exhibit neuroprotective effects. Large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels are key modulators of cellular excitability of neurons and vascular smooth muscle cells. In the present study, we examined the effects of ginsenosides on rat brain BK(Ca) (rSlo) channel activity heterologously expressed in Xenopus oocytes to elucidate the molecular mechanisms how ginsenoside regulates the BK(Ca) channel activity. Ginsenoside Rg(3) (Rg(3)) enhanced outward BK(Ca) channel currents. The Rg(3)-enhancement of outward BK(Ca) channel currents was concentration-dependent, voltage-dependent, and reversible. The EC(50) was 15.1 ± 3.1 μM. Rg(3) actions were not desensitized by repeated treatment. Tetraetylammonium (TEA), a K(+) channel blocker, inhibited BK(Ca) channel currents. We examined whether extracellular TEA treatment could alter the Rg(3) action and vice versa. TEA caused a rightward shift of the Rg(3) concentration-response curve (i.e., much higher concentration of Rg(3) is required for the activation of BK(Ca) channel compared to the absence of TEA), while Rg(3) caused a rightward shift of the TEA concentration-response curve in wild-type channels. Mutation of the extracellular TEA binding site Y360 to Y360I caused a rightward shift of the TEA concentration-response curve and almost abolished both the Rg(3) action and Rg(3)-induced rightward shift of TEA concentration-response curve. These results indicate that Tyr360 residue of BK(Ca) channel plays an important role in the Rg(3)-enhancement of BK(Ca) channel currents.
Collapse
Affiliation(s)
- Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 143-701, Korea
| | - Tae-Joon Shin
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 143-701, Korea
| | - Byung-Hwan Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 143-701, Korea
| | - Sung Hee Hwang
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 143-701, Korea
| | - Sang-Mok Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 143-701, Korea
| | - Byung-Cheol Lee
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Cheol-Seung Park
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Tal Soo Ha
- Department of Molecular Biology, College of Natural Science, Daegu University, Gyeongsan 712-714, Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul 143-701, Korea
| |
Collapse
|
10
|
Flagg TP, Enkvetchakul D, Koster JC, Nichols CG. Muscle KATP channels: recent insights to energy sensing and myoprotection. Physiol Rev 2010; 90:799-829. [PMID: 20664073 DOI: 10.1152/physrev.00027.2009] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
ATP-sensitive potassium (K(ATP)) channels are present in the surface and internal membranes of cardiac, skeletal, and smooth muscle cells and provide a unique feedback between muscle cell metabolism and electrical activity. In so doing, they can play an important role in the control of contractility, particularly when cellular energetics are compromised, protecting the tissue against calcium overload and fiber damage, but the cost of this protection may be enhanced arrhythmic activity. Generated as complexes of Kir6.1 or Kir6.2 pore-forming subunits with regulatory sulfonylurea receptor subunits, SUR1 or SUR2, the differential assembly of K(ATP) channels in different tissues gives rise to tissue-specific physiological and pharmacological regulation, and hence to the tissue-specific pharmacological control of contractility. The last 10 years have provided insights into the regulation and role of muscle K(ATP) channels, in large part driven by studies of mice in which the protein determinants of channel activity have been deleted or modified. As yet, few human diseases have been correlated with altered muscle K(ATP) activity, but genetically modified animals give important insights to likely pathological roles of aberrant channel activity in different muscle types.
Collapse
Affiliation(s)
- Thomas P Flagg
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
11
|
Rollo CD. Dopamine and Aging: Intersecting Facets. Neurochem Res 2008; 34:601-29. [DOI: 10.1007/s11064-008-9858-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Accepted: 07/03/2008] [Indexed: 10/21/2022]
|
12
|
Wang G, Pan J, Tan YY, Sun XK, Zhang YF, Zhou HY, Ren RJ, Wang XJ, Chen SD. Neuroprotective effects of PACAP27 in mice model of Parkinson's disease involved in the modulation of K(ATP) subunits and D2 receptors in the striatum. Neuropeptides 2008; 42:267-76. [PMID: 18440632 DOI: 10.1016/j.npep.2008.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 02/24/2008] [Accepted: 03/05/2008] [Indexed: 10/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) exhibits a protective effect against neural injury in vitro and in vivo. However, it has not been reported whether peripheral intravenous administration of PACAP could confer benefits in animal models of Parkinson's disease (PD). Furthermore, the underlying molecular mechanisms responsible for these effects are poorly understood. In the present experiments, we determined the effects and mechanism of action of intravenously administered PACAP27 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Our results indicate that intravenous injection of PACAP27 offers neuroprotective effects in the MPTP-induced PD mouse model which may not be directly associated with the expression levels of the monoamine transporters. However, this effect may be correlated with its ability to selectively regulate not only K(ATP) subunits, but D2 receptors in the striatum. Our findings suggest that the benefit of PACAP may accompany with changes not only in dopaminergic neurotransmission, but also in cholinergic neurotransmission that are relatively associated with the K(ATP) subunits and D2 receptors in the striatum.
Collapse
Affiliation(s)
- Gang Wang
- Department of Neurology and Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Pacheco Otalora LF, Hernandez EF, Arshadmansab MF, rancisco SF, Willis M, Ermolinsky B, Zarei M, Knaus HG, Garrido-Sanabria ER. Down-regulation of BK channel expression in the pilocarpine model of temporal lobe epilepsy. Brain Res 2008; 1200:116-31. [PMID: 18295190 PMCID: PMC2346580 DOI: 10.1016/j.brainres.2008.01.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2007] [Revised: 12/23/2007] [Accepted: 01/03/2008] [Indexed: 11/24/2022]
Abstract
In the hippocampus, BK channels are preferentially localized in presynaptic glutamatergic terminals including mossy fibers where they are thought to play an important role regulating excessive glutamate release during hyperactive states. Large conductance calcium-activated potassium channels (BK, MaxiK, Slo) have recently been implicated in the pathogenesis of genetic epilepsy. However, the role of BK channels in acquired mesial temporal lobe epilepsy (MTLE) remains unknown. Here we used immunohistochemistry, laser scanning confocal microscopy (LSCM), Western immunoblotting and RT-PCR to investigate the expression pattern of the alpha-pore-forming subunit of BK channels in the hippocampus and cortex of chronically epileptic rats obtained by the pilocarpine model of MTLE. All epileptic rats experiencing recurrent spontaneous seizures exhibited a significant down-regulation of BK channel immunostaining in the mossy fibers at the hilus and stratum lucidum of the CA3 area. Quantitative analysis of immunofluorescence signals by LSCM revealed a significant 47% reduction in BK channel immunofluorescent signals in epileptic rats when compared to age-matched non-epileptic control rats. These data correlate with a similar reduction in BK channel protein levels and transcripts in the cortex and hippocampus. Our data indicate a seizure-related down-regulation of BK channels in chronically epileptic rats. Further functional assays are necessary to determine whether altered BK channel expression is an acquired channelopathy or a compensatory mechanism affecting the network excitability in MTLE. Moreover, seizure-mediated BK down-regulation may disturb neuronal excitability and presynaptic control at glutamatergic terminals triggering exaggerated glutamate release and seizures.
Collapse
Affiliation(s)
- Luis F. Pacheco Otalora
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
| | - Eder F. Hernandez
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
| | - Massoud F. Arshadmansab
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
| | - Sebastian F rancisco
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
| | - Michael Willis
- Department of General Psychiatry, Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
- Department of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter-Mayr Strasse 1, 6020 Innsbruck, Austria
| | - Boris Ermolinsky
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
| | - Masoud Zarei
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
- The Center for Biomedical Studies, Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Hans-Guenther Knaus
- Department of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter-Mayr Strasse 1, 6020 Innsbruck, Austria
| | - Emilio R. Garrido-Sanabria
- Department of Biological Sciences at the University of Texas at Brownsville/Texas Southmost College, Brownsville, Texas 78520 USA
- The Center for Biomedical Studies, Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
14
|
Abstract
The ATP-sensitive potassium (K(ATP)) channels which extensively distribute in diverse tissues (e.g. vascular smooth muscle, cardiac cells, and pancreas) are well-established for characteristics like vasodilatation, myocardial protection against ischemia, and insulin secretion. The aim of this review is to get insight into the novel roles of K(ATP) channels in Parkinson's disease (PD), with consideration of the specificities K(ATP) channels in the central nervous system (CNS), such as the control of neuronal excitability, action potential, mitochondrial function and neurotransmitter release.
Collapse
|
15
|
Abstract
BACKGROUND AND PURPOSE Sulphatides are sulphated glycosphingolipids expressed on the surface of many cell types, particularly neurones. Changes in sulphatide species or content have been associated with epilepsy and Alzheimer's disease. As the large conductance, calcium sensitive K(+) channel (BK(Ca)) are modulated by membrane lipids, the aim of the study was to explore possible effects of sulphatides on BK(Ca) channels. EXPERIMENTAL APPROACH Using patch-clamp techniques, we studied effects of exogenous sulphatides on BK(Ca) channels expressed in Chinese hamster ovary cells. KEY RESULTS Sulphatides reversibly increased the whole-cell current and the single channel open probability of BK(Ca) channels dose-dependently. The EC(50) value on the channel at +10 mV was 1.6 microM and the Hill coefficient was 2.5. In inside-out patches, sulphatides increased the single channel open probability from both intra- and extra-cellular faces of the membrane, but more effectively with external application. Furthermore, activation of the channels by sulphatides was independent of intracellular Ca(2+) concentration. Sulphatides also shifted the activation curve of the channels to less positive membrane potentials. Mutant BK(Ca) channels lacking a 59 aminoacid region important for amphipath activation (STREX) were less activated by the sulphatides. CONCLUSIONS AND IMPLICATIONS Sulphatides are novel activators of BK(Ca) channels, independent of intracellular Ca(2+) or other signalling molecules but partly dependent on the STREX sequence of the channel protein. As changes of sulphatide content are associated with neuronal dysfunction, as in epilepsy and Alzheimer's disease, our results imply that these effects of sulphatides may play important pathophysiological roles in regulation of BK(Ca) channels.
Collapse
Affiliation(s)
- S Chi
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics Beijing, PR China
| | - Z Qi
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics Beijing, PR China
- Author for correspondence:
| |
Collapse
|
16
|
Richter A, Sander SE, Rundfeldt C. Antidystonic effects of Kv7 (KCNQ) channel openers in the dt sz mutant, an animal model of primary paroxysmal dystonia. Br J Pharmacol 2006; 149:747-53. [PMID: 17016514 PMCID: PMC2014660 DOI: 10.1038/sj.bjp.0706878] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Mutations in neuronal Kv7 (KCNQ) potassium channels can cause episodic neurological disorders. Paroxysmal dyskinesias with dystonia are a group of movement disorders which are regarded as ion channelopathies, but the role of Kv7 channels in the pathogenesis and as targets for the treatment have so far not been examined. EXPERIMENTAL APPROACH In the present study, we therefore examined the effects of the activators of neuronal Kv7.2/7.3 channels retigabine (5, 7.5, 10 mg kg(-1) i.p. and 10, 20 mg kg(-1) p.o.) and flupirtine (10, 20 mg kg(-1) i.p.) and of the channel blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone (XE-991, 3 and 6 mg kg(-1) i.p.) in the dt sz mutant hamster, a model of paroxysmal dyskinesia in which dystonic episodes occur in response to stress. KEY RESULTS Retigabine (10 mg kg(-1) i.p., 20 mg kg(-1) p.o.) and flupirtine (20 mg kg(-1) i.p.) significantly improved dystonia, while XE-991 caused a significant aggravation in the dt sz mutant. The antidystonic effect of retigabine (10 mg kg(-1) i.p.) was counteracted by XE-991 (3 mg kg(-1) i.p.). CONCLUSIONS AND IMPLICATIONS These data indicate that dysfunctions of neuronal Kv7 channels deserve attention in dyskinesias. Since retigabine and flupirtine are well tolerated in humans, the present finding of pronounced antidystonic efficacy in the dt sz mutant suggests that neuronal Kv7 channel activators are interesting candidates for the treatment of dystonia-associated dyskinesias and probably of other types of dystonias. The established analgesic effects of Kv7 channel openers might contribute to improvement of these disorders which are often accompanied by painful muscle spasms.
Collapse
Affiliation(s)
- A Richter
- Institute of Pharmacology and Toxicology, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| | | | | |
Collapse
|
17
|
Morimura K, Yamamura H, Ohya S, Imaizumi Y. Voltage-dependent Ca2+-channel block by openers of intermediate and small conductance Ca2+-activated K+ channels in urinary bladder smooth muscle cells. J Pharmacol Sci 2006; 100:237-41. [PMID: 16518073 DOI: 10.1254/jphs.sc0060011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
We examined effects of small and intermediate conductance Ca(2+)-activated K(+) (SK and IK) channel openers, DCEBIO (5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one) and NS309 (3-oxime-6,7-dichloro-1H-indole-2,3-dione), on L-type Ca(2+) channel current (I(Ca)) that was measured in smooth muscle cells isolated from mouse urinary bladder under whole cell voltage-clamp. The I(Ca) was concentration-dependently inhibited by DCEBIO and NS309; half inhibition was obtained at 71.6 and 10.6 muM, respectively. The specificity of NS309 to the IK channel over the Ca(2+) channel appears to be high and higher than that of DCEBIO. DCEBIO and even NS309 may, however, substantially block Ca(2+) channels when used as SK channel openers.
Collapse
Affiliation(s)
- Kozo Morimura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | | | | | | |
Collapse
|
18
|
Zhang L, Li X, Zhou R, Xing G. Possible role of potassium channel, big K in etiology of schizophrenia. Med Hypotheses 2006; 67:41-3. [PMID: 16446048 DOI: 10.1016/j.mehy.2005.09.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Accepted: 09/29/2005] [Indexed: 11/17/2022]
Abstract
Schizophrenia (SZ), a common severe mental disorder, affecting about 1% of the world population. However, the etiology of SZ is still largely unknown. It is believed that molecules that are in an association with the etiology and pathology of SZ are neurotransmitters including dopamine, 5-HT and gamma-aminobutyric acid (GABA). But several lines of evidences indicate that potassium large conductance calcium-activated channel, known as BK channel, is likely to be included. BK channel belongs to a group of ion channels that plays an important role in regulating neuronal excitability and transmitter releasing. Its involvement in SZ emerges as a great interest. For example, commonly used neuroleptics, in clinical therapeutic concentrations, alter calcium-activated potassium conductance in central neurons. Diazoxide, a potassium channel opener/activator, showed a significant superiority over haloperidol alone in the treatment of positive and general psychopathology symptoms in SZ. Additionally, estrogen, which regulates the activity of BK channel, modulates dopaminergic D2 receptor and has an antipsychotic-like effect. Therefore, we hypothesize that BK channel may play a role in SZ and those agents, which can target either BK channel functions or its expression may contribute to the therapeutic actions of SZ treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Psychiatry, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814-4799, United States.
| | | | | | | |
Collapse
|
19
|
Sade H, Muraki K, Ohya S, Hatano N, Imaizumi Y. Activation of large-conductance, Ca2+-activated K+channels by cannabinoids. Am J Physiol Cell Physiol 2006; 290:C77-86. [PMID: 16107501 DOI: 10.1152/ajpcell.00482.2004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We have examined the effects of the cannabinoid anandamide (AEA) and its stable analog, methanandamide (methAEA), on large-conductance, Ca2+-activated K+(BK) channels using human embryonic kidney (HEK)-293 cells, in which the α-subunit of the BK channel (BK-α), both α- and β1-subunits (BK-αβ1), or both α- and β4-subunits (BK-αβ4) were heterologously expressed. In a whole cell voltage-clamp configuration, each cannabinoid activated BK-αβ1within a similar concentration range. Because methAEA could potentiate BK-α, BK-αβ1, and BK-αβ4with similar efficacy, the β-subunits may not be involved at the site of action for cannabinoids. Under cell-attached patch-clamp conditions, application of methAEA to the bathing solution increased BK channel activity; however, methAEA did not alter channel activity in the excised inside-out patch mode even when ATP was present on the cytoplasmic side of the membrane. Application of methAEA to HEK-BK-α and HEK-BK-αβ1did not change intracellular Ca2+concentration. Moreover, methAEA-induced potentiation of BK channel currents was not affected by pretreatment with a CB1antagonist (AM251), modulators of G proteins (cholera and pertussis toxins) or by application of a selective CB2agonist (JWH133). Inhibitors of CaM, PKG, and MAPKs (W7, KT5823, and PD-98059) did not affect the potentiation. Application of methAEA to mouse aortic myocytes significantly increased BK channel currents. This study provides the first direct evidence that unknown factors in the cytoplasm mediate the ability of endogenous cannabinoids to activate BK channel currents. Cannabinoids may be hyperpolarizing factors in cells, such as arterial myocytes, in which BK channels are highly expressed.
Collapse
Affiliation(s)
- Hiroko Sade
- Dept. of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City Univ., 3-1 Tanabedori, Mizuhoku, Nagoya 467-8603, Japan
| | | | | | | | | |
Collapse
|
20
|
Ha TS, Lim HH, Lee GE, Kim YC, Park CS. Electrophysiological Characterization of Benzofuroindole-Induced Potentiation of Large-Conductance Ca2+-Activated K+ Channels. Mol Pharmacol 2005; 69:1007-14. [PMID: 16332986 DOI: 10.1124/mol.105.016170] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Large-conductance Ca2+-activated K+ (BK(Ca)) channels are widely distributed and play key roles in various cell functions. We previously reported the chemical synthesis of several benzofuroindole compounds that act as potent openers of BK(Ca) channels. In this study, we investigated the mechanism of channel potentiation by one of the compounds, 7-trifluoromethyl-10H-benzo[4,5]furo[3,2-b]indole-1-carboxylic acid (TBIC), using electrophysiological means. This chemical highly activated cloned BK(Ca) channels from extracellular side independent of beta subunits and regardless of the presence of intracellular Ca2+. The EC50 and Hill coefficient for rat BK(Ca) channel alpha subunit, rSlo, were estimated as 8.9 +/- 1.5 microM and 0.9, respectively. TBIC shifted the conductance-voltage curve of rSlo channels to more hyperpolarized potentials without altering its voltage dependence. Single-channel recording revealed that TBIC increased the open probability of the channel in a dose-dependent manner without any changes in single-channel conductance. Strong potentiation by TBIC was also observed for native BK(Ca) channels from rat hippocampus pyramidal neurons. Thus, TBIC and the related benzofuroindole compounds can be useful tools to unravel the mechanism of this novel allosteric activation of BK(Ca) channels.
Collapse
Affiliation(s)
- Tal Soo Ha
- Department of Life Science, Gwangju Institute of Science and Technology, Bukgu, Gwangju, 500-712, Korea
| | | | | | | | | |
Collapse
|
21
|
Myslobodsky M. Phobic memory and somatic vulnerabilities in anorexia nervosa: a necessary unity? Ann Gen Psychiatry 2005; 4:15. [PMID: 16144551 PMCID: PMC1260012 DOI: 10.1186/1744-859x-4-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2005] [Accepted: 09/06/2005] [Indexed: 11/10/2022] Open
Abstract
Anorexia nervosa is a clinically significant illness that may be associated with permanent medical complications involving almost every organ system. The paper raises a question whether some of them are associated with premorbid vulnerability such as subcellular ion channel abnormalities ('channelopathy') that determines the clinical expression of the bodily response to self-imposed malnutrition. Aberrant channels emerge as a tempting, if rather speculative alternative to the notion of cognitively-driven neurotransmitter modulation deficit in anorexia nervosa. The concept of channelopathies is in keeping with some characteristics of anorexia nervosa, such as a genetically-based predisposition to hypophagia, early onset, cardiac abnormalities, an appetite-enhancing efficacy of some antiepileptic drugs, and others. The purpose of this article is to stimulate further basic research of ion channel biophysics in relation to restrictive anorexia.
Collapse
|
22
|
Abstract
Ion channels are membrane proteins that flicker open and shut to regulate the flow of ions down their electrochemical gradient across the membrane and consequently regulate cellular excitability. Every living cell expresses ion channels, as they are critical life-sustaining proteins. Ion channels are generally either activated by voltage or by ligand interaction. For each group of ion channels the channels' molecular biology and biophysics will be introduced and the pharmacology of that group of channels will be reviewed. The in vitro and in vivo literature will be reviewed and, for ion channel groups in which clinical trials have been conducted, the efficacy and therapeutic potential of the neuroprotective compounds will be reviewed. A large part of this article will deal with glutamate receptors, focusing specifically on N-methyl-D-aspartate (NMDA) receptors. Although the outcome of clinical trials for NMDA receptor antagonists as therapeutics for acute stroke is disappointing, the culmination of these failed trials was preceded by a decade of efforts to develop these agents. Sodium and calcium channel antagonists will be reviewed and the newly emerging efforts to develop therapeutics targeting potassium channels will be discussed. The future development of stroke therapeutics targeting ion channels will be discussed in the context of the failures of the last decade in hopes that this decade will yield successful stroke therapeutics.
Collapse
Affiliation(s)
- D L Small
- Institute or Biological Sciences, National Research Council of Canada, Building M-54, 1200 Montreal Road, Ottawa, Ontario, Canada K1A 0R6.
| |
Collapse
|
23
|
Sarpal D, Koenig JI, Adelman JP, Brady D, Prendeville LC, Shepard PD. Regional distribution of SK3 mRNA-containing neurons in the adult and adolescent rat ventral midbrain and their relationship to dopamine-containing cells. Synapse 2004; 53:104-13. [PMID: 15170822 DOI: 10.1002/syn.20042] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
SK3 small conductance, calcium-activated potassium channels play an important role in regulating the activity of mesencephalic dopamine (DA) neurons. In the present series of experiments, in situ hybridization techniques were used to compare SK3 and tyrosine hydroxylase (TH) mRNA expression throughout the rostrocaudal extent of the ventral midbrain in juvenile and adult rats. SK3 mRNA was found exclusively in areas that also contained large numbers of DA neurons including the substantia nigra (SN), the ventral tegmental area, and related cell groups (VTA-A10). An anteroposterior and mediolateral gradient in SK3 mRNA hybridization was apparent in the VTA-A10 but not in the SN. Younger rats appeared to possess higher levels and less regional variation in TH and SK3 transcripts. These results are consistent with previous studies reporting differential expression of SK3 protein within the midbrain and suggest that variations in SK3 channel distribution could contribute to differences in dopamine-related functions in the rat.
Collapse
Affiliation(s)
- Deepak Sarpal
- Maryland Psychiatric Research Center, Department of Psychiatry and Program in Neuroscience, University of Maryland School of Medicine, Baltimore 21228, USA
| | | | | | | | | | | |
Collapse
|
24
|
Mannhold R. KATP channel openers: structure-activity relationships and therapeutic potential. Med Res Rev 2004; 24:213-66. [PMID: 14705169 DOI: 10.1002/med.10060] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
ATP-sensitive potassium channels (K(ATP) channels) are heteromeric complexes of pore-forming inwardly rectifying potassium channel subunits and regulatory sulfonylurea receptor subunits. K(ATP) channels were identified in a variety of tissues including muscle cells, pancreatic beta-cells, and various neurons. They are regulated by the intracellular ATP/ADP ratio; ATP induces channel inhibition and MgADP induces channel opening. Functionally, K(ATP) channels provide a means of linking the electrical activity of a cell to its metabolic state. Shortening of the cardiac action potential, smooth muscle relaxation, inhibition of both insulin secretion, and neurotransmitter release are mediated via K(ATP) channels. Given their many physiological functions, K(ATP) channels represent promising drug targets. Sulfonylureas like glibenclamide block K(ATP) channels; they are used in the therapy of type 2 diabetes. Openers of K(ATP) channels (KCOs), for example, relax smooth muscle and induce hypotension. KCOs are chemically heterogeneous and include as different classes as the benzopyrans, cyanoguanidines, thioformamides, thiadiazines, and pyridyl nitrates. Examples for new chemical entities more recently developed as KCOs include cyclobutenediones, dihydropyridine related structures, and tertiary carbinols.
Collapse
Affiliation(s)
- Raimund Mannhold
- Department of Laser Medicine, Molecular Drug Research Group, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
25
|
Kwon S, Lee S, Hyun M, Choe BH, Kim Y, Park W, Cho Y. The potential for QT prolongation by antiepileptic drugs in children. Pediatr Neurol 2004; 30:99-101. [PMID: 14984900 DOI: 10.1016/s0887-8994(03)00405-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2003] [Accepted: 07/02/2003] [Indexed: 10/26/2022]
Abstract
Cardiac arrhythmia may be one of the major causes of sudden unexpected death in children with epilepsy. We assessed drug-induced QT prolongation to establish whether the use of antiepileptic drugs contributes to sudden unexpected death. A total of 178 children with epilepsy (93 males and 85 females, with ages ranging from 1 month to 18.9 years; mean age 7.0 +/- 4.1 years) were involved in the study. The QT intervals were manually measured and corrected using Fridericia's formula (QTFc = QT/RR(1/3)). The mean corrected QT interval (QTc) of 152 children on antiepileptic drugs during the study period was 0.40 +/- 0.03 s, and for 26 age-matched, antiepileptic drug-free control patients it was 0.40 +/- 0.03 s. The mean QTc of the children with monotherapy was 0.40 +/- 0.03 s for the valproate group (n = 42), 0.39 +/- 0.02 s for the carbamazepine/oxcarbazepine group (n = 34), and 0.40 +/- 0.02 s for the topiramate group (n = 26), respectively. There was no statistically significant difference among the groups as assessed by analysis of variance. In addition, there was no significant difference between the monotherapy group (n = 109; 0.40 +/- 0.02 s) and the polytherapy group (n = 43; 0.39 +/- 0.03 s). Major antiepileptic drugs may not precipitate prolongation of the QT interval into sudden unexpected death in children with epilepsy, however further studies are required.
Collapse
Affiliation(s)
- Soonhak Kwon
- Department of Pediatrics, Kyungpook National University Hospital, Joong-gu, Daegu, South Korea
| | | | | | | | | | | | | |
Collapse
|
26
|
Postlethwaite M, Constanti A. Evidence for the involvement of G-proteins in the generation of the slow poststimulus afterdepolarisation (sADP) induced by muscarinic receptor activation in rat olfactory cortical neurones in vitro. Brain Res 2003; 978:124-35. [PMID: 12834906 DOI: 10.1016/s0006-8993(03)02799-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The involvement of G-proteins in generating the slow poststimulus afterdepolarising potential (sADP) induced by muscarinic receptor activation in immature (P10-20) rat olfactory cortical brain slice neurones was investigated under whole-cell patch clamp, using GTP-gamma-S (G-protein activator) or GDP-beta-S (G-protein blocker)-filled electrodes. In control experiments using K methylsulphate electrodes, cell resting potential (V(m)) and spike firing properties were unaffected over 10-15 min recording, although input resistance (R(N)) was slightly increased ( approximately 14%). Oxotremorine-M (OXO-M; 10 microM) produced a reversible slow depolarisation, an increase in R(N) ( approximately 90%) and induction of a slow poststimulus inward tail current (I(ADP)) (measured under voltage clamp at -60 mV) that was sustained during drug exposure (up to 15 min); the amplitude of slow inward rectifier (I(h)) currents activated from -50 mV were also apparently increased. By contrast, in GTP-gamma-S-loaded cells, R(N) was consistently decreased ( approximately 22%) and spike firing threshold (V(th)) was raised ( approximately 5 mV) after 10 min recording. In approximately 60% of loaded cells, a persistent muscarinic slow inward current and I(ADP) were induced by OXO-M; I(h) relaxation amplitude was also significantly decreased. The effects of GTP-gamma-S on R(N), V(th) and I(h) were partly counteracted by adding Ba(2+) (100 microM) to the bathing medium or mimicked by adding baclofen (GABA(B) receptor agonist; 100 microM) to normally-recorded cells. Intracellular GDP-beta-S (up to 30 min) had no effect on cell membrane properties or I(h), but irreversibly blocked the muscarinic slow inward current and I(ADP) induced by OXO-M. We conclude that both muscarinic responses require G-protein-linked transduction mechanisms for their generation.
Collapse
Affiliation(s)
- Michael Postlethwaite
- Department of Pharmacology, The School of Pharmacy, 29/39 Brunswick Square, London WC1N 1AX, UK
| | | |
Collapse
|
27
|
Imaizumi Y, Sakamoto K, Yamada A, Hotta A, Ohya S, Muraki K, Uchiyama M, Ohwada T. Molecular basis of pimarane compounds as novel activators of large-conductance Ca(2+)-activated K(+) channel alpha-subunit. Mol Pharmacol 2002; 62:836-46. [PMID: 12237330 DOI: 10.1124/mol.62.4.836] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Effects of pimaric acid (PiMA) and eight closely related compounds on large-conductance K(+) (BK) channels were examined using human embryonic kidney (HEK) 293 cells, in which either the alpha subunit of BK channel (HEKBKalpha) or both alpha and beta1 (HEKBKalphabeta1) subunits were heterologously expressed. Effects of these compounds (10 microM) on the membrane potential of HEKBKalphabeta1 were monitored by use of DiBAC(4)(3), a voltage-sensitive dye. PiMA, isopimaric acid, sandaracoisopimaric acid, dihydropimaric acid, dihydroisopimaric acid, and dihydroisopimarinol induced substantial membrane hyperpolarization. The direct measurement of BKalphabeta1 opening under whole-cell voltage clamp showed that these six compounds activated BKalphabeta1 in a very similar concentration range (1-10 microM); in contrast, abietic acid, sclareol, and methyl pimarate had no effect. PiMA did not affect the charybdotoxin-induced block of macroscopic BKalphabeta1 current. Single channel recordings of BKalphabeta1 in inside-out patches showed that 10 microM PiMA did not change channel conductance but significantly increased its open probability as a result of increase in sensitivity to Ca(2+) and voltage. Because coexpression of the beta1 subunit did not affect PiMA-induced potentiation, the site of action for PiMA is suggested to be BKalpha subunit. PiMA was selective to BK over cloned small and intermediate Ca(2+) activated K(+) channels. In conclusion, PiMA (>1 microM) increases Ca(2+) and voltage-sensitivity of BKalpha when applied from either side of the cell membrane. The marked difference in potency as BK channel openers between PiMA and abietic acid, despite only very small differences in their chemical structures, may provide insight into the fundamental structure-activity relationship governing BKalpha activation.
Collapse
Affiliation(s)
- Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Lawson K. Tricyclic antidepressants and fibromyalgia: what is the mechanism of action? Expert Opin Investig Drugs 2002; 11:1437-45. [PMID: 12387704 DOI: 10.1517/13543784.11.10.1437] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibromyalgia is a chronic pain disorder of which other clinical features, such as persistent fatigue and disordered sleep, may be a secondary consequence. The initial pharmacological approach to treating the disorder is the management of the pain. Tricyclic antidepressants are the most effective drugs in use so far, especially when administered in combination with other therapies (e.g., selective serotonin re-uptake inhibitors), which suggests modulation of the neurotransmitters serotonin and noradrenaline. The effectiveness of amitriptyline and related tricyclic antidepressants, however, is consistent with the involvement of mechanisms, such as potassium channel modulation and NMDA receptor antagonism, in addition to or in place of the modulation of monoamine neurotransmitters. Investigation of the importance of each of the pharmacological properties of amitriptyline and related molecules in the management of fibromyalgia could provide clues for the rational design of new drugs.
Collapse
Affiliation(s)
- Kim Lawson
- Biomedical Research Centre and Division of Biomedical Sciences, Sheffield Hallam University, School of Science and Mathematics, City Campus, UK.
| |
Collapse
|
29
|
Yamada A, Gaja N, Ohya S, Muraki K, Narita H, Ohwada T, Imaizumi Y. Usefulness and limitation of DiBAC4(3), a voltage-sensitive fluorescent dye, for the measurement of membrane potentials regulated by recombinant large conductance Ca2+-activated K+ channels in HEK293 cells. JAPANESE JOURNAL OF PHARMACOLOGY 2001; 86:342-50. [PMID: 11488436 DOI: 10.1254/jjp.86.342] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The usefulness of bis-(1,3-dibutylbarbituric acid)-trimethine oxonol (DiBAC4(3)), a voltage-sensitive fluorescent dye, for the measurement of membrane potentials (MPs) was evaluated in HEK293 cells, where alpha or alpha plus beta1 subunits of large conductance Ca2+-activated K+ (BK) channels were expressed (HEKBK alpha and HEKBK alphabeta). The fluorescent intensity of DiBAC4(3) was measured at various potentials under voltage-clamp for calibration to estimate the absolute MP semi-quantitatively. The resting MPs measured with DiBAC4(3) were roughly comparable to those recorded with a microelectrode; the MP in HEKBK alphabeta was 10-20 mV more negative than that in native HEK. In HEKBK alpha, the membrane hyperpolarization induced by 10 microM Evans blue, a BK channel opener, was detected with DiBAC4(3). NS-1619, another BK channel opener, induced gradual but substantial change in F/F(K) even in native HEK, while the BK channel opening effect was detected. Oscillatory membrane hyperpolarization was induced in HEKBK alphabeta by application of 10 microM acetylcholine via increase in intracellular Ca2+ concentration. The oscillatory hyperpolarization was, however, detected only as a slow hyperpolarization with DiBAC4(3). It can be concluded that relatively slow effects of BK channel modulators can be semi-quantitatively measured by use of DiBAC4(3) in HEKBK, while the limited temporal resolution and possible artifacts should be taken into account.
Collapse
Affiliation(s)
- A Yamada
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|