1
|
Tsvetkov D, Schleifenbaum J, Wang Y, Kassmann M, Polovitskaya MM, Ali M, Schütze S, Rothe M, Luft FC, Jentsch TJ, Gollasch M. KCNQ5 Controls Perivascular Adipose Tissue-Mediated Vasodilation. Hypertension 2024; 81:561-571. [PMID: 38354270 DOI: 10.1161/hypertensionaha.123.21834] [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: 07/24/2023] [Accepted: 12/12/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND Small arteries exhibit resting tone, a partially contracted state that maintains arterial blood pressure. In arterial smooth muscle cells, potassium channels control contraction and relaxation. Perivascular adipose tissue (PVAT) has been shown to exert anticontractile effects on the blood vessels. However, the mechanisms by which PVAT signals small arteries, and their relevance remain largely unknown. We aimed to uncover key molecular components in adipose-vascular coupling. METHODS A wide spectrum of genetic mouse models targeting Kcnq3, Kcnq4, and Kcnq5 genes (Kcnq3-/-, Kcnq4-/-, Kcnq5-/-, Kcnq5dn/dn, Kcnq4-/-/Kcnq5dn/dn, and Kcnq4-/-/Kcnq5-/-), telemetry blood pressure measurements, targeted lipidomics, RNA-Seq profiling, wire-myography, patch-clamp, and sharp-electrode membrane potential measurements was used. RESULTS We show that PVAT causes smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels to hyperpolarize the membrane potential. This effect relaxes small arteries and regulates blood pressure. Oxygenation of polyunsaturated fats generates oxylipins, a superclass of lipid mediators. We identified numerous oxylipins released by PVAT, which potentiate vasodilatory action in small arteries by opening smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels. CONCLUSIONS Our results reveal a key molecular function of the KV7.5 family of voltage-gated potassium (K+) channels in the adipose-vascular coupling, translating PVAT signals, particularly oxylipins, to the central physiological function of vasoregulation. This novel pathway opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| | - Johanna Schleifenbaum
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Germany (J.S.)
| | - Yibin Wang
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany (Y.W., F.C.L.)
| | - Mario Kassmann
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| | - Maya M Polovitskaya
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (M.M.P., S.S., T.J.J.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (M.M.P., S.S., T.J.J.)
| | - Mohamed Ali
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| | - Sebastian Schütze
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (M.M.P., S.S., T.J.J.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (M.M.P., S.S., T.J.J.)
| | | | - Friedrich C Luft
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany (Y.W., F.C.L.)
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (M.M.P., S.S., T.J.J.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (M.M.P., S.S., T.J.J.)
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany (T.J.J.)
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| |
Collapse
|
2
|
Qian X, Zhao X, Yu L, Yin Y, Zhang XD, Wang L, Li JX, Zhu Q, Luo JL. Current status of GABA receptor subtypes in analgesia. Biomed Pharmacother 2023; 168:115800. [PMID: 37935070 DOI: 10.1016/j.biopha.2023.115800] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
Gamma-aminobutyric acid (GABA), a non-protein-producing amino acid synthesized from the excitatory amino acid glutamate via the enzyme glutamic acid decarboxylase, is extensively found in microorganisms, plants and vertebrates, and is abundantly expressed in the spinal cord and brain. It is the major inhibitory neurotransmitter in the mammalian nervous system. GABA plays crucial roles in the regulation of synaptic transmission, the promotion of neuronal development and relaxation, and the prevention of insomnia and depression. As the major inhibitory neurotransmitter, GABA plays pivotal roles in the regulation of pain sensation, which is initiated by the activation of peripheral nociceptors and transmitted to the spinal cord and brain along nerves. GABA exerts these roles by directly acting on three types of receptors: ionotropic GABAA and GABAC receptors and G protein-coupled GABAB receptor. The chloride-permeable ion channel receptors GABAA and GABAC mediate fast neurotransmission, while the metabotropic GABAB receptor mediates slow effect. Different GABA receptors regulate pain sensation via different signaling pathways. Here we highlight recent updates on the involvement of specific GABA receptors and their subtypes in the process of pain sensation. Further understanding of different GABA receptors and signaling pathways in pain sensation will benefit the development of novel analgesics for pain management by targeting specific GABA receptor subtypes and signaling pathways.
Collapse
Affiliation(s)
- Xunjia Qian
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Xinyi Zhao
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Lulu Yu
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yujian Yin
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Xiao-Dan Zhang
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Liyun Wang
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jun-Xu Li
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong 226001, Jiangsu, China.
| | - Jia-Lie Luo
- School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
3
|
Kir Channel Molecular Physiology, Pharmacology, and Therapeutic Implications. Handb Exp Pharmacol 2021; 267:277-356. [PMID: 34345939 DOI: 10.1007/164_2021_501] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
For the past two decades several scholarly reviews have appeared on the inwardly rectifying potassium (Kir) channels. We would like to highlight two efforts in particular, which have provided comprehensive reviews of the literature up to 2010 (Hibino et al., Physiol Rev 90(1):291-366, 2010; Stanfield et al., Rev Physiol Biochem Pharmacol 145:47-179, 2002). In the past decade, great insights into the 3-D atomic resolution structures of Kir channels have begun to provide the molecular basis for their functional properties. More recently, computational studies are beginning to close the time domain gap between in silico dynamic and patch-clamp functional studies. The pharmacology of these channels has also been expanding and the dynamic structural studies provide hope that we are heading toward successful structure-based drug design for this family of K+ channels. In the present review we focus on placing the physiology and pharmacology of this K+ channel family in the context of atomic resolution structures and in providing a glimpse of the promising future of therapeutic opportunities.
Collapse
|
4
|
Nickerson AJ, Rajendran VM. Flupirtine enhances NHE-3-mediated Na + absorption in rat colon via an ENS-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 2021; 321:G185-G199. [PMID: 34132108 PMCID: PMC8410105 DOI: 10.1152/ajpgi.00158.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 01/31/2023]
Abstract
Recent studies in our lab have shown that the KV7 channel activator, flupirtine, inhibits colonic epithelial Cl- secretion through effects on submucosal neurons of the enteric nervous system (ENS). We hypothesized that flupirtine would also stimulate Na+ absorption as a result of reduced secretory ENS input to the epithelium. To test this hypothesis, unidirectional 22Na+ fluxes were measured under voltage-clamped conditions. Pharmacological approaches using an Ussing-style recording chamber combined with immunofluorescence microscopy techniques were used to determine the effect of flupirtine on active Na+ transport in the rat colon. Flupirtine stimulated electroneutral Na+ absorption in partially seromuscular-stripped colonic tissues, while simultaneously inhibiting short-circuit current (ISC; i.e., Cl- secretion). Both of these effects were attenuated by pretreatment with the ENS inhibitor, tetrodotoxin. The Na+/H+ exchanger isoform 3 (NHE-3)-selective inhibitor, S3226, significantly inhibited flupirtine-stimulated Na+ absorption, whereas the NHE-2-selective inhibitor HOE-694 did not. NHE-3 localization near the apical membranes of surface epithelial cells was also more apparent in flupirtine-treated colon versus control. Flupirtine did not alter epithelial Na+ channel (ENaC)-mediated Na+ absorption in distal colonic tissues obtained from hyperaldosteronaemic rats and had no effect in the normal ileum but did stimulate Na+ absorption in the proximal colon. Finally, the parallel effects of flupirtine on ISC (Cl- secretion) and Na+ absorption were significantly correlated with each other. Together, these data indicate that flupirtine stimulates NHE-3-dependent Na+ absorption, likely as a result of reduced stimulatory input to the colonic epithelium by submucosal ENS neurons.NEW & NOTEWORTHY We present a novel mechanism regarding regulation of epithelial ion transport by enteric neurons. Activation of neuronal KV7 K+ channels markedly stimulates Na+ absorption and inhibits Cl- secretion across the colonic epithelium. This may be useful in developing new treatments for diarrheal disorders, such as irritable bowel syndrome with diarrhea (IBS-D).
Collapse
Affiliation(s)
- Andrew J Nickerson
- Departments of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
5
|
Failed, Interrupted, or Inconclusive Trials on Neuroprotective and Neuroregenerative Treatment Strategies in Multiple Sclerosis: Update 2015-2020. Drugs 2021; 81:1031-1063. [PMID: 34086251 PMCID: PMC8217012 DOI: 10.1007/s40265-021-01526-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
In the recent past, a plethora of drugs have been approved for the treatment of multiple sclerosis (MS). These therapeutics are mainly confined to immunomodulatory or immunosuppressive strategies but do not sufficiently address remyelination and neuroprotection. However, several neuroregenerative agents have shown potential in pre-clinical research and entered Phase I to III clinical trials. Although none of these compounds have yet proceeded to approval, understanding the causes of failure can broaden our knowledge about neuroprotection and neuroregeneration in MS. Moreover, most of the investigated approaches are characterised by consistent mechanisms of action and proved convincing efficacy in animal studies. Therefore, learning from their failure will help us to enforce the translation of findings acquired in pre-clinical studies into clinical application. Here, we summarise trials on MS treatment published since 2015 that have either failed or were interrupted due to a lack of efficacy, adverse events, or for other reasons. We further outline the rationale underlying these drugs and analyse the background of failure to gather new insights into MS pathophysiology and optimise future study designs. For conciseness, this review focuses on agents promoting remyelination and medications with primarily neuroprotective properties or unconventional approaches. Failed clinical trials that pursue immunomodulation are presented in a separate article.
Collapse
|
6
|
Nickerson AJ, Rottgen TS, Rajendran VM. Activation of KCNQ (K V7) K + channels in enteric neurons inhibits epithelial Cl - secretion in mouse distal colon. Am J Physiol Cell Physiol 2021; 320:C1074-C1087. [PMID: 33852365 PMCID: PMC8285638 DOI: 10.1152/ajpcell.00536.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Voltage-gated Kv7 (KCNQ family) K+ channels are expressed in many neuronal populations and play an important role in regulating membrane potential by generating a hyperpolarizing K+ current and decreasing cell excitability. However, the role of KV7 channels in the neural regulation of intestinal epithelial Cl- secretion is not known. Cl- secretion in mouse distal colon was measured as a function of short-circuit current (ISC), and pharmacological approaches were used to test the hypothesis that activation of KV7 channels in enteric neurons would inhibit epithelial Cl- secretion. Flupirtine, a nonselective KV7 activator, inhibited basal Cl- secretion in mouse distal colon and abolished or attenuated the effects of drugs that target various components of enteric neurotransmission, including tetrodotoxin (NaV channel blocker), veratridine (NaV channel activator), nicotine (nicotinic acetylcholine receptor agonist), and hexamethonium (nicotinic antagonist). In contrast, flupritine did not block the response to epithelium-targeted agents VIP (endogenous VPAC receptor ligand) or carbachol (nonselective cholinergic agonist). Flupirtine inhibited Cl- secretion in both full-thickness and seromuscular-stripped distal colon (containing the submucosal, but not myenteric plexus) but generated no response in epithelial T84 cell monolayers. KV7.2 and KV7.3 channel proteins were detected by immunofluorescence in whole mount preparations of the submucosa from mouse distal colon. ICA 110381 (KV7.2/7.3 specific activator) inhibited Cl- secretion comparably to flupirtine. We conclude that KV7 channel activators inhibit neurally driven Cl- secretion in the colonic epithelium and may therefore have therapeutic benefit in treating pathologies associated with hyperexcitable enteric nervous system, such as irritable bowel syndrome with diarrhea (IBS-D).
Collapse
Affiliation(s)
- Andrew J Nickerson
- Departments of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Trey S Rottgen
- Departments of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
7
|
Borgini M, Mondal P, Liu R, Wipf P. Chemical modulation of Kv7 potassium channels. RSC Med Chem 2021; 12:483-537. [PMID: 34046626 PMCID: PMC8128042 DOI: 10.1039/d0md00328j] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/01/2020] [Indexed: 01/10/2023] Open
Abstract
The rising interest in Kv7 modulators originates from their ability to evoke fundamental electrophysiological perturbations in a tissue-specific manner. A large number of therapeutic applications are, in part, based on the clinical experience with two broad-spectrum Kv7 agonists, flupirtine and retigabine. Since precise molecular structures of human Kv7 channel subtypes in closed and open states have only very recently started to emerge, computational studies have traditionally been used to analyze binding modes and direct the development of more potent and selective Kv7 modulators with improved safety profiles. Herein, the synthetic and medicinal chemistry of small molecule modulators and the representative biological properties are summarized. Furthermore, new therapeutic applications supported by in vitro and in vivo assay data are suggested.
Collapse
Affiliation(s)
- Matteo Borgini
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Pravat Mondal
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Ruiting Liu
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| |
Collapse
|
8
|
Lawson K. Pharmacology and clinical applications of flupirtine: Current and future options. World J Pharmacol 2019; 8:1-13. [DOI: 10.5497/wjp.v8.i1.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/17/2018] [Accepted: 01/05/2019] [Indexed: 02/06/2023] Open
Abstract
Flupirtine is the first representative in a class of triaminopyridines that exhibits pharmacological properties leading to the suppression of over-excitability of neuronal and non-neuronal cells. Consequently, this drug has been used as a centrally acting analgesic in patients with a range of acute and persistent pain conditions without the adverse effects characteristic of opioids and non-steroidal anti-inflammatory drug and is well tolerated. The pharmacological profile exhibited involves actions on several cellular targets, including Kv7 channels, G-protein-regulated inwardly rectifying K channels and γ-aminobutyric acid type A receptors, but also there is evidence of additional as yet unidentified mechanisms of action involved in the effects of flupirtine. Flupirtine has exhibited effects in a range of cells and tissues related to the locations of these targets. In additional to analgesia, flupirtine has demonstrated pharmacological properties consistent with use as an anticonvulsant, a neuroprotectant, skeletal and smooth muscle relaxant, in treatment of auditory and visual disorders, and treatment of memory and cognitive impairment. Flupirtine is providing important information and clues regarding novel mechanistic approaches to the treatment of a range of clinical conditions involving hyper-excitability of cells. Identification of molecules exhibiting specificity for the pharmacological targets (e.g., Kv7 isoforms) involved in the actions of flupirtine will provide further insight into clinical applications. Whether the broad-spectrum pharmacology of flupirtine or target-specific actions is preferential to gain benefit, especially in complex clinical conditions, requires further investigation. This review will consider recent advancement in understanding of the pharmacological profile and related clinical applications of flupirtine.
Collapse
Affiliation(s)
- Kim Lawson
- Department of Biosciences and Chemistry, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, United Kingdom
| |
Collapse
|
9
|
Patil MA, Matter BA, Raol YH, Bourne DWA, Kelley RA, Kompella UB. Brain Distribution and Metabolism of Flupirtine, a Nonopioid Analgesic Drug with Antiseizure Effects, in Neonatal Rats. Pharmaceutics 2018; 10:E281. [PMID: 30558371 PMCID: PMC6320943 DOI: 10.3390/pharmaceutics10040281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
Flupirtine, a nonopioid analgesic drug, is effective in treating neonatal seizures. However, its brain delivery and pharmacokinetics are unknown in neonatal mammals. The purpose of this study was to determine the pharmacokinetics of flupirtine and the formation of its active metabolite D-13223 in various tissues such as brain in neonate animals. On postnatal day 7, rat pups received 25 mg/kg of flupirtine intraperitoneally. Liver; heart; kidney; lung; spleen; retina; serum; and brain regions hippocampus, cortex, and the remaining brain (devoid of cerebellum) were harvested up to 24-h postdosing. An LC-MS/MS assay was developed to quantify flupirtine and D-13223. Flupirtine was delivered to all tissues assessed, with the highest area under the concentration vs. time curve (AUC0⁻24h) in liver (488 µg·h/g tissue) and the lowest in spleen (82 µg·h/g tissue). Flupirtine reached the brain, including the hippocampus and cortex, within 1 h of dosing and persisted at 24 h. Flupirtine AUC in various brain regions was approximately 195 µg·h/g tissue. The half-life of flupirtine in various tissues ranged from 3.1 to 5.2 h. D-13223 was formed in vivo and detected in all tissues assessed, with the concentrations being the highest in the liver. Incubation of isolated neonatal rat liver, heart, kidney, lung, spleen, whole eye, serum, or whole brain with flupirtine for 3 h at 37 °C formed D-13223 in all tissues, except serum. D-13223 formation was the highest in isolated liver tissue. Tissue partition coefficients based on isolated tissue uptake correlated well with in vivo tissue:serum drug exposure ratios. Thus, flupirtine reaches the target brain tissues from the systemic route in neonatal rats, and brain tissue forms the active metabolite D-13223.
Collapse
Affiliation(s)
- Madhoosudan A Patil
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Brock A Matter
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Yogendra H Raol
- Department of Pediatrics, Division of Neurology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - David W A Bourne
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Ryan A Kelley
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Uday B Kompella
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
10
|
Sampath D, Valdez R, White AM, Raol YH. Anticonvulsant effect of flupirtine in an animal model of neonatal hypoxic-ischemic encephalopathy. Neuropharmacology 2017; 123:126-135. [PMID: 28587899 DOI: 10.1016/j.neuropharm.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/07/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022]
Abstract
Research studies suggest that neonatal seizures, which are most commonly associated with hypoxic-ischemic injury, may contribute to brain injury and adverse neurologic outcome. Unfortunately, neonatal seizures are often resistant to treatment with current anticonvulsants. In the present study, we evaluated the efficacy of flupirtine, administered at clinically relevant time-points, for the treatment of neonatal seizures in an animal model of hypoxic-ischemic injury that closely replicates features of the human syndrome. We also compared the efficacy of flupirtine to that of phenobarbital, the current first-line drug for neonatal seizures. Flupirtine is a KCNQ potassium channel opener. KCNQ channels play an important role in controlling brain excitability during early development. In this study, hypoxic-ischemic injury was induced in neonatal rats, and synchronized video-EEG records were acquired at various time-points during the experiment to identify seizures. The results revealed that flupirtine, administered either 5 min after the first electroclinical seizure, or following completion of 2 h of hypoxia, i.e., during the immediate reperfusion period, reduced the number of rats with electroclinical seizures, and also the frequency and total duration of electroclinical seizures. Further, daily dosing of flupirtine decreased the seizure burden over 3 days following HI-induction, and modified the natural evolution of acute seizures. Moreover, compared to a therapeutic dose of phenobarbital, which was modestly effective against electroclinical seizures, flupirtine showed greater efficacy. Our results indicate that flupirtine is an extremely effective treatment for neonatal seizures in rats and provide evidence for a trial of this medication in newborn humans.
Collapse
Affiliation(s)
- Dayalan Sampath
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert Valdez
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andrew M White
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yogendra H Raol
- Department of Pediatrics, Division of Neurology, School of Medicine, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
11
|
Yadav G, Jain G, Singh M. Role of flupirtine in reducing preoperative anxiety of patients undergoing craniotomy procedure. Saudi J Anaesth 2017; 11:158-162. [PMID: 28442953 PMCID: PMC5389233 DOI: 10.4103/1658-354x.203028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Kv7 neuronal channels are recognized as a potential drug target for anxiolytic effects. We hypothesize that flupirtine as a potassium channel opener would effectively reduce the preoperative anxiety of patients undergoing craniotomy procedure. Methods: In prospective-double-blinded fashion, 124 counseled patients were randomized to receive 5 sequential doses of capsule flupirtine 100 mg (F Group) or physically similar starch capsules (C Group), at 12 h intervals during preoperative hospitalization. Primary outcome included various aspects of patient anxiety measured by visual analog scale (VAS) just before preoperative counseling and 2 h after the completion of drug regimen under trial. Statistical tool included Mann–Whitney U-test and Wilcoxon signed rank test. Results: Baseline VAS scores were higher for fear of surgical harm, being at the mercy of medical staff, and not awakening after surgery. A significant decline in VAS scores was observed after the completion of drug regime, but to a higher extent in flupirtine-treated patients; it achieved statistical significance in comparison to Group C. No side effects were observed in any patient. Conclusion: Flupirtine is a useful premedication in conjunction with behavioral therapy to alleviate patient anxiety during the preoperative period.
Collapse
Affiliation(s)
- Ghanshyam Yadav
- Department of Anaesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Gaurav Jain
- Department of Anaesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Malkhan Singh
- Department of Anaesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
12
|
Jaeger HM, Pehlke JR, Kaltwasser B, Kilic E, Bähr M, Hermann DM, Doeppner TR. The indirect NMDAR inhibitor flupirtine induces sustained post-ischemic recovery, neuroprotection and angioneurogenesis. Oncotarget 2016; 6:14033-44. [PMID: 26050199 PMCID: PMC4546449 DOI: 10.18632/oncotarget.4226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/13/2015] [Indexed: 12/20/2022] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) activation induces excitotoxicity, contributing to post-stroke brain injury. Hitherto, NMDAR deactivation failed in clinical trials due to insufficient pre-clinical study designs and drug toxicity. Flupirtine is an indirect NMDAR antagonist being used as analgesic in patients. Taking into account its tolerability profile, we evaluated effects of flupirtine on post-stroke tissue survival, neurological recovery and brain remodeling. Mice were exposed to stroke and intraperitoneally treated with saline (control) or flupirtine at various doses (1-10 mg/kg) and time-points (0-12 hours). Tissue survival and cell signaling were studied on day 2, whereas neurological recovery and tissue remodeling were analyzed until day 84. Flupirtine induced sustained neuroprotection, when delivered up to 9 hours. The latter yielded enhanced neurological recovery that persisted over three months and which was accompanied by enhanced angioneurogenesis. On the molecular level, inhibition of calpain activation was noted, which was associated with increased signal-transducer-and-activator-of-transcription-6 (STAT6) abundance, reduced N-terminal-Jun-kinase and NF-κB activation, as well as reduced proteasomal activity. Consequently, blood-brain-barrier integrity was stabilized, oxidative stress was reduced and brain leukocyte infiltration was diminished. In view of its excellent tolerability, considering its sustained effects on neurological recovery, brain tissue survival and remodeling, flupirtine is an attractive candidate for stroke therapy.
Collapse
Affiliation(s)
- Hanna M Jaeger
- University of Duisburg-Essen Medical School, Department of Neurology, Essen, Germany
| | - Jens R Pehlke
- LWL-Klinik Muenster, Department of Addiction Disorders, Muenster, Germany
| | - Britta Kaltwasser
- University of Duisburg-Essen Medical School, Department of Neurology, Essen, Germany
| | - Ertugrul Kilic
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey
| | - Mathias Bähr
- University of Goettingen Medical School, Department of Neurology, Goettingen, Germany
| | - Dirk M Hermann
- University of Duisburg-Essen Medical School, Department of Neurology, Essen, Germany
| | - Thorsten R Doeppner
- University of Duisburg-Essen Medical School, Department of Neurology, Essen, Germany.,Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey
| |
Collapse
|
13
|
Klinger F, Bajric M, Salzer I, Dorostkar MM, Khan D, Pollak DD, Kubista H, Boehm S, Koenig X. δ Subunit-containing GABAA receptors are preferred targets for the centrally acting analgesic flupirtine. Br J Pharmacol 2015. [PMID: 26211808 PMCID: PMC4621994 DOI: 10.1111/bph.13262] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background and Purpose The Kv7 channel activator flupirtine is a clinical analgesic characterized as ‘selective neuronal potassium channel opener’. Flupirtine was found to exert comparable actions at GABAA receptors and Kv7 channels in neurons of pain pathways, but not in hippocampus. Experimental Approach Expression patterns of GABAA receptors were explored in immunoblots of rat dorsal root ganglia, dorsal horns and hippocampi using antibodies for 10 different subunits. Effects of flupirtine on recombinant and native GABAA receptors were investigated in patch clamp experiments and compared with the actions on Kv7 channels. Key Results Immunoblots pointed towards α2, α3, β3 and γ2 subunits as targets, but in all γ2‐containing receptors the effects of flupirtine were alike: leftward shift of GABA concentration‐response curves and diminished maximal amplitudes. After replacement of γ2S by δ, flupirtine increased maximal amplitudes. Currents through α1β2δ receptors were more enhanced than those through Kv7 channels. In hippocampal neurons, flupirtine prolonged inhibitory postsynaptic currents, left miniature inhibitory postsynaptic currents (mIPSCs) unaltered and increased bicuculline‐sensitive tonic currents; penicillin abolished mIPSCs, but not tonic currents; concentration‐response curves for GABA‐induced currents were shifted to the left by flupirtine without changes in maximal amplitudes; in the presence of penicillin, maximal amplitudes were increased; GABA‐induced currents in the presence of penicillin were more sensitive towards flupirtine than K+ currents. In dorsal horn neurons, currents evoked by the δ‐preferring agonist THIP (gaboxadol) were more sensitive towards flupirtine than K+ currents. Conclusions and Implications Flupirtine prefers δ‐containing GABAA receptors over γ‐containing ones and over Kv7 channels.
Collapse
Affiliation(s)
- Felicia Klinger
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Mirnes Bajric
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Mario M Dorostkar
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Deeba Khan
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Helmut Kubista
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| | - Xaver Koenig
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Austria
| |
Collapse
|
14
|
Synergistic interaction between tapentadol and flupirtine in the rat orafacial formalin test. Eur J Pharmacol 2015; 762:350-6. [PMID: 26048311 DOI: 10.1016/j.ejphar.2015.05.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/26/2015] [Accepted: 05/29/2015] [Indexed: 12/17/2022]
Abstract
Combination therapy with two or more analgesics is widely used for conditions associated with moderate to severe pain. Combinations of diverse analgesics with different modes of action can improve the risk-benefit ratio of analgesic treatments. The aim of this study is to evaluate the antinociceptive effect of tapentadol (TAP) and flupirtine (FLP), when administered separately or in combination, as well as their synergistic interaction in the orofacial formalin test in rats. After i.p. injection of TAP at different doses (2, 5, 10 and 15mg/kg), the biphasic nociceptive behavior was reduced in a dose-dependent manner in both phase I and II. Conversely, i.p. injection of FLP at different doses (0.6, 1.6, 3.3, 6.6, 16.6 and 22.2mg/kg) induced a dose-dependent antinociceptive effect in phase II only. TAP was found to be more effective than FLP. The interaction between TAP and FLP was synergistic in phase II with an interaction index (γ) of 0.50±0.24. The data reported in this study indicate that FLP enhances the antinociceptive effect of TAP and this drug combination might be potentially useful in the treatment of chronic pain.
Collapse
|
15
|
Yadav G, Behera SS, Das SK, Jain G, Choupoo S, Raj J. Role of flupirtine as a preemptive analgesic in patients undergoing laparoscopic cholecystectomy. J Anaesthesiol Clin Pharmacol 2015; 31:169-73. [PMID: 25948895 PMCID: PMC4411828 DOI: 10.4103/0970-9185.155143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background and Aims: Postsurgical pain is the leading complaint after laparoscopic cholecystectomy that may delay the postoperative recovery and hence we undertook a prospective randomized trial to analyze the role of flupirtine as a preemptive analgesic for postoperative pain relief in patients undergoing above surgery. Material and Methods: A total of 66 cases were randomly assigned to two groups to receive capsule flupirtine (200 mg) or capsule vitamin B complex administered orally, 2 h before the laparoscopic cholecystectomy surgery. Time to first analgesic requirement, assessment of postoperative pain in terms of visual analog score, and analgesic requirement postoperatively were measured as a primary outcome. Results: Time to first analgesic requirement was significantly prolonged in the flupirtine group as compared with the placebo group. There was significant pain reduction in early postoperative period (up to 4 h), but no changes occurred thereafter. Total analgesic requirement (including rescue analgesia) and side-effects were comparable between the groups except for higher sedation in flupirtine group. Conclusions: Flupirtine is effective as a preemptive analgesic in providing adequate pain relief during the immediate postoperative period after laparoscopic cholecystectomy surgery. However, continuation of drug therapy postoperatively could possibly delineate its optimal analgesic profile more profoundly.
Collapse
Affiliation(s)
- Ghanshyam Yadav
- Department of Anesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Shailaja Shankar Behera
- Department of Anesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Saurabh Kumar Das
- Department of Anesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Gaurav Jain
- Department of Anesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sujali Choupoo
- Department of Anesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Janak Raj
- Department of Neurosurgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
16
|
Wörz R. [Long-term-treatment of chronic pain patients with flupirtine--on hepatotoxicity and persistent effectiveness from 7 months to 22 years]. MMW Fortschr Med 2014; 156 Suppl 4:127-134. [PMID: 26153592 DOI: 10.1007/s15006-014-3759-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
BACKGROUND After an extended series of investigations in animals, healthy volunteers and pain patients with different methods, flupirtine was introduced in 1986 into the German market. Flupirtine has a unique chemical structure and differs in its profile of effects and side effects from established NSAIDs (non-steroidal anti-inflammatory drugs) on the one hand and from opioids on the other hand. In the years after introduction, muscle relaxing effects were observed and experimentally shown. They were therapeutically utilized in tension-associated pain. Evidence of the efficacy in acute and chronic musculoskeletal pain syndromes was verified in many controlled and in open studies. METHOD Experience report and case presentation RESULTS The report in this paper of long-term treatment of chronic pain patients from 7 months to 22 years principally confirms its persistent effectiveness and tolerability. Tiredness and dizziness are frequent side effects. Flupirtine-associated hepatotoxicities are rare events. CONCLUSION For pain patients for which NSAIDs or opioids are contraindicated or in appropriate, flupirtine is a necessary option.
Collapse
|
17
|
Borin M, Fogli Iseppe A, Pignatelli A, Belluzzi O. Inward rectifier potassium (Kir) current in dopaminergic periglomerular neurons of the mouse olfactory bulb. Front Cell Neurosci 2014; 8:223. [PMID: 25152712 PMCID: PMC4126183 DOI: 10.3389/fncel.2014.00223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 07/21/2014] [Indexed: 11/23/2022] Open
Abstract
Dopaminergic (DA) periglomerular (PG) neurons are critically placed at the entry of the bulbar circuitry, directly in contact with both the terminals of olfactory sensory neurons and the apical dendrites of projection neurons; they are autorhythmic and are the target of numerous terminals releasing a variety of neurotransmitters. Despite the centrality of their position, suggesting a critical role in the sensory processing, their properties -and consequently their function- remain elusive. The current mediated by inward rectifier potassium (Kir) channels in DA-PG cells was recorded by adopting the perforated-patch configuration in thin slices; IKir could be distinguished from the hyperpolarization-activated current (I h ) by showing full activation in <10 ms, no inactivation, suppression by Ba(2+) in a typical voltage-dependent manner (IC50 208 μM) and reversal potential nearly coincident with EK. Ba(2+) (2 mM) induces a large depolarization of DA-PG cells, paralleled by an increase of the input resistance, leading to a block of the spontaneous activity, but the Kir current is not an essential component of the pacemaker machinery. The Kir current is negatively modulated by intracellular cAMP, as shown by a decrease of its amplitude induced by forskolin or 8Br-cAMP. We have also tested the neuromodulatory effects of the activation of several metabotropic receptors known to be present on these cells, showing that the current can be modulated by a multiplicity of pathways, whose activation in some case increases the amplitude of the current, as can be observed with agonists of D2, muscarinic, and GABAA receptors, whereas in other cases has the opposite effect, as it can be observed with agonists of α1 noradrenergic, 5-HT and histamine receptors. These characteristics of the Kir currents provide the basis for an unexpected plasticity of DA-PG cell function, making them potentially capable to reconfigure the bulbar network to allow a better flexibility.
Collapse
Affiliation(s)
| | | | | | - Ottorino Belluzzi
- Department of Life Sciences and Biotechnology, University of FerraraFerrara, Italy
| |
Collapse
|
18
|
Wu C, V Gopal K, Lukas TJ, Gross GW, Moore EJ. Pharmacodynamics of potassium channel openers in cultured neuronal networks. Eur J Pharmacol 2014; 732:68-75. [PMID: 24681057 DOI: 10.1016/j.ejphar.2014.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 03/06/2014] [Accepted: 03/17/2014] [Indexed: 12/30/2022]
Abstract
A novel class of drugs - potassium (K(+)) channel openers or activators - has recently been shown to cause anticonvulsive and neuroprotective effects by activating hyperpolarizing K(+) currents, and therefore, may show efficacy for treating tinnitus. This study presents measurements of the modulatory effects of four K(+) channel openers on the spontaneous activity and action potential waveforms of neuronal networks. The networks were derived from mouse embryonic auditory cortices and grown on microelectrode arrays. Pentylenetetrazol was used to create hyperactivity states in the neuronal networks as a first approximation for mimicking tinnitus or tinnitus-like activity. We then compared the pharmacodynamics of the four channel activators, retigabine and flupirtine (voltage-gated K(+) channel KV7 activators), NS1619 and isopimaric acid ("big potassium" BK channel activators). The EC50 of retigabine, flupirtine, NS1619, and isopimaric acid were 8.0, 4.0, 5.8, and 7.8µM, respectively. The reduction of hyperactivity compared to the reference activity was significant. The present results highlight the notion of re-purposing the K(+) channel activators for reducing hyperactivity of spontaneously active auditory networks, serving as a platform for these drugs to show efficacy toward target identification, prevention, as well as treatment of tinnitus.
Collapse
Affiliation(s)
- Calvin Wu
- Department of Speech and Hearing Sciences, University of North Texas, Denton, TX 76203, United States; Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States; Center for Network Neuroscience, University of North Texas, Denton, TX 76203, United States.
| | - Kamakshi V Gopal
- Department of Speech and Hearing Sciences, University of North Texas, Denton, TX 76203, United States; Center for Network Neuroscience, University of North Texas, Denton, TX 76203, United States
| | - Thomas J Lukas
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Guenter W Gross
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, United States; Center for Network Neuroscience, University of North Texas, Denton, TX 76203, United States
| | - Ernest J Moore
- Department of Speech and Hearing Sciences, University of North Texas, Denton, TX 76203, United States; Center for Network Neuroscience, University of North Texas, Denton, TX 76203, United States; Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| |
Collapse
|
19
|
Abstract
Cell shrinkage is a hallmark and contributes to signaling of apoptosis. Apoptotic cell shrinkage requires ion transport across the cell membrane involving K(+) channels, Cl(-) or anion channels, Na(+)/H(+) exchange, Na(+),K(+),Cl(-) cotransport, and Na(+)/K(+)ATPase. Activation of K(+) channels fosters K(+) exit with decrease of cytosolic K(+) concentration, activation of anion channels triggers exit of Cl(-), organic osmolytes, and HCO3(-). Cellular loss of K(+) and organic osmolytes as well as cytosolic acidification favor apoptosis. Ca(2+) entry through Ca(2+)-permeable cation channels may result in apoptosis by affecting mitochondrial integrity, stimulating proteinases, inducing cell shrinkage due to activation of Ca(2+)-sensitive K(+) channels, and triggering cell-membrane scrambling. Signaling involved in the modification of cell-volume regulatory ion transport during apoptosis include mitogen-activated kinases p38, JNK, ERK1/2, MEKK1, MKK4, the small G proteins Cdc42, and/or Rac and the transcription factor p53. Osmosensing involves integrin receptors, focal adhesion kinases, and tyrosine kinase receptors. Hyperosmotic shock leads to vesicular acidification followed by activation of acid sphingomyelinase, ceramide formation, release of reactive oxygen species, activation of the tyrosine kinase Yes with subsequent stimulation of CD95 trafficking to the cell membrane. Apoptosis is counteracted by mechanisms involved in regulatory volume increase (RVI), by organic osmolytes, by focal adhesion kinase, and by heat-shock proteins. Clearly, our knowledge on the interplay between cell-volume regulatory mechanisms and suicidal cell death is still far from complete and substantial additional experimental effort is needed to elucidate the role of cell-volume regulatory mechanisms in suicidal cell death.
Collapse
Affiliation(s)
- Florian Lang
- Institute of Physiology, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
20
|
Gahr M, Freudenmann RW, Connemann BJ, Hiemke C, Schönfeldt-Lecuona C. Abuse liability of flupirtine revisited: Implications of spontaneous reports of adverse drug reactions. J Clin Pharmacol 2013; 53:1328-33. [DOI: 10.1002/jcph.164] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/13/2013] [Indexed: 01/28/2023]
Affiliation(s)
- Maximilian Gahr
- Department of Psychiatry and Psychotherapy III; University Hospital of Ulm; Ulm Germany
| | - Roland W. Freudenmann
- Department of Psychiatry and Psychotherapy III; University Hospital of Ulm; Ulm Germany
| | - Bernhard J. Connemann
- Department of Psychiatry and Psychotherapy III; University Hospital of Ulm; Ulm Germany
| | - Christoph Hiemke
- Department of Psychiatry and Psychotherapy; University Medical Center of Mainz; Mainz Germany
| | | |
Collapse
|
21
|
Gahr M, Freudenmann RW, Kölle MA, Schönfeldt-Lecuona C. Dependence on flupirtine. J Clin Pharmacol 2013; 53:1003-4. [PMID: 23846806 DOI: 10.1002/jcph.129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 06/04/2013] [Indexed: 12/17/2022]
|
22
|
Noam Y, Raol YH, Holmes GL. Searching for new targets for treatment of pediatric epilepsy. Epilepsy Behav 2013; 26:253-60. [PMID: 23219411 PMCID: PMC3595393 DOI: 10.1016/j.yebeh.2012.09.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 09/06/2012] [Indexed: 12/13/2022]
Abstract
The highest incidence of seizures in humans occurs during the first year of life. The high susceptibility to seizures in neonates and infants is paralleled by animal studies showing a high propensity to seizures during early life. The immature brain is highly susceptible to seizures because of an imbalance of excitation and inhibition. While the primary outcome determinant of early-life seizures is etiology, there is evidence that seizures which are frequent or prolonged can result in long-term adverse consequences, and there is a consensus that recurrent early-life seizures should be treated. Unfortunately, seizures in many neonates and children remain refractory to therapy. There is therefore a pressing need for new seizure drugs as well as antiepileptic targets in children. In this review, we focus on mechanisms of early-life seizures, such as hypoxia-ischemia, and novel molecular targets, including the hyperpolarization-activated cyclic nucleotide-gated channels.
Collapse
Affiliation(s)
- Yoav Noam
- Department of Anatomy & Neurobiology, University of California-Irvine, Irvine, California
| | - Yogendra H. Raol
- Division of Neurology, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Gregory L. Holmes
- Department of Neurology Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
23
|
Flupirtine, a re-discovered drug, revisited. Inflamm Res 2013; 62:251-8. [PMID: 23322112 DOI: 10.1007/s00011-013-0592-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/20/2012] [Accepted: 01/02/2013] [Indexed: 12/15/2022] Open
Abstract
Flupirtine was developed long before K(V)7 (KCNQ) channels were known. However, it was clear from the beginning that flupirtine is neither an opioid nor a nonsteroidal anti-inflammatory analgesic. Its unique muscle relaxing activity was discovered by serendipity. In the meantime, broad and intensive research has resulted in a partial clarification of its mode of action. Flupirtine is the first therapeutically used K(V)7 channel activator with additional GABA(A)ergic mechanisms and thus the first representative of a novel class of analgesics. The presently accepted main mode of its action, potassium K(V)7 (KCNQ) channel activation, opens a series of further therapeutic possibilities. One of them has now been realized: its back-up compound, the bioisostere retigabine, has been approved for the treatment of epilepsy.
Collapse
|
24
|
Klinger F, Geier P, Dorostkar MM, Chandaka GK, Yousuf A, Salzer I, Kubista H, Boehm S. Concomitant facilitation of GABAA receptors and KV7 channels by the non-opioid analgesic flupirtine. Br J Pharmacol 2012; 166:1631-42. [PMID: 22188423 DOI: 10.1111/j.1476-5381.2011.01821.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Flupirtine is a non-opioid analgesic that has been in clinical use for more than 20 years. It is characterized as a selective neuronal potassium channel opener (SNEPCO). Nevertheless, its mechanisms of action remain controversial and are the purpose of this study. EXPERIMENTAL APPROACH Effects of flupirtine on native and recombinant voltage- and ligand-gated ion channels were explored in patch-clamp experiments using the following experimental systems: recombinant K(IR)3 and K(V)7 channels and α3β4 nicotinic acetylcholine receptors expressed in tsA 201 cells; native voltage-gated Na(+), Ca(2+), inward rectifier K(+), K(V)7 K(+), and TRPV1 channels, as well as GABA(A), glycine, and ionotropic glutamate receptors expressed in rat dorsal root ganglion, dorsal horn and hippocampal neurons. KEY RESULTS Therapeutic flupirtine concentrations (≤10 µM) did not affect voltage-gated Na(+) or Ca(2+) channels, inward rectifier K(+) channels, nicotinic acetylcholine receptors, glycine or ionotropic glutamate receptors. Flupirtine shifted the gating of K(V)7 K(+) channels to more negative potentials and the gating of GABA(A) receptors to lower GABA concentrations. These latter effects were more pronounced in dorsal root ganglion and dorsal horn neurons than in hippocampal neurons. In dorsal root ganglion and dorsal horn neurons, the facilitatory effect of therapeutic flupirtine concentrations on K(V)7 channels and GABA(A) receptors was comparable, whereas in hippocampal neurons the effects on K(V)7 channels were more pronounced. CONCLUSIONS AND IMPLICATIONS These results indicate that flupirtine exerts its analgesic action by acting on both GABA(A) receptors and K(V)7 channels.
Collapse
Affiliation(s)
- Felicia Klinger
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Flupirtine is neither an opioid nor a non steroidal anti-inflammatory drug (NSAID) producing its analgesic action through blockade of glutamate N-methyl-D-aspartate receptor. It is devoid of adverse effects of routinely used analgesic drugs, but is equally efficacious in reducing pain sensation. It has a distinctive mechanism of action, exerting a dual therapeutic effect with both analgesic and muscle relaxant properties that has utility in the treatment of pain, including that associated with muscle tension.
Collapse
Affiliation(s)
- S Harish
- Department of Pharmacology, Sri Devaraj Urs Medical College, Tamaka, Kolar, Karnataka, India
| | | | | | | |
Collapse
|
26
|
Uberall MA, Mueller-Schwefe GHH, Terhaag B. Efficacy and safety of flupirtine modified release for the management of moderate to severe chronic low back pain: results of SUPREME, a prospective randomized, double-blind, placebo- and active-controlled parallel-group phase IV study. Curr Med Res Opin 2012; 28:1617-34. [PMID: 22970658 DOI: 10.1185/03007995.2012.726216] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To demonstrate non-inferior/superior efficacy of flupirtine modified release (MR) compared with tramadol/placebo for the management of moderate to severe chronic low back pain (LBP). RESEARCH DESIGN Randomized, double-blind, active-/placebo-controlled double-dummy multicenter study, performed in 31 German study centers. LBP patients (n = 363) with moderate pain intensity were randomized 1:1:1 to receive flupirtine MR 400 mg, tramadol extended release (ER) 200 mg, or matching placebo (each given OD in the evening) over 4 weeks. CLINICAL TRIAL REGISTRATION EudraCT 2009-013268-38. MAIN OUTCOME MEASURES Primary endpoint was change from baseline in the LBP intensity index (LBPIX; 11-point NRS) at week 4; last observation carried forward was used to impute missing scores. RESULTS Least square (LS) mean ± SD LBPIX changes from baseline at week 4 were clinically significant for all three treatment groups of the intent-to-treat (ITT) and the per-protocol (PP) population (n = 326/276): placebo (n = 110/96): -1.81 ± 1.65/-1.77 ± 1.59; flupirtine MR (n = 109/95): -2.23 ± 1.73/-2.28 ± 1.68; and tramadol ER (n = 107/85): -1.92 ± 1.84/2.03 ± 1.83 (p < 0.001 for each). ITT/PP treatment effects for flupirtine MR were non-inferior when compared with tramadol ER and superior when compared with placebo (p = 0.003/0.033). Significantly more ITT patients treated with flupirtine MR (59.6/37.6 showed a ≥30/50% LBPIX relief in comparison to placebo (46.4/24.6%; p vs. flupirtine MR: 0.049/0.037). Treatment contrasts for tramadol failed to reach significance vs. placebo. Within the safety population (n = 355), flupirtine MR (n = 119) was associated with a significantly lower incidence of treatment emergent AEs (TEAEs; 21.0%) and TEAE-related study discontinuations (3.4%) than tramadol ER (n = 116; 34.5/12.0%; p = 0.039/0.017) and exhibited an overall safety/tolerability profile non-inferior to placebo (n = 120; 15.8/3.3%; p = ns for each). Major limitations of this study were the short treatment duration, the comparison of different drug classes and the lack of a titration phase. CONCLUSIONS The analgesic efficacy of flupirtine MR 400 mg OD was comparable to that of tramadol ER 200 mg OD and superior to that of placebo.
Collapse
Affiliation(s)
- Michael A Uberall
- Institute for Neurological Sciences, Algesiology and Pediatrics, Nuernberg, Germany.
| | | | | |
Collapse
|
27
|
Singal R, Gupta P, Jain N, Gupta S. Role of flupirtine in the treatment of pain - chemistry and its effects. MAEDICA 2012; 7:163-166. [PMID: 23401726 PMCID: PMC3557425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 05/05/2012] [Indexed: 06/01/2023]
Affiliation(s)
- Rikki Singal
- Department of Surgery - Maharishi Markandeshwer Institute of Medical Sciences and Research, Mullana, (Distt-Ambala), Haryana, India E
| | | | | | | |
Collapse
|
28
|
Abstract
BACKGROUND Perivascular adipose tissue secretes an adipocyte-derived relaxing factor (ADRF) that opens voltage-dependent K (Kv) channels in peripheral arteries. We studied the role of KCNQ-type Kv channels and tested the hypothesis that hydrogen sulfide (H2S) could be an ADRF. METHODS We performed isometric contraction studies on systemic arteries of rats and mice. RESULTS In mesenteric arteries and aortas without perivascular adipose tissue, the KCNQ channel openers retigabine, VRX0530727, VRX0621238, and VRX0621688 produced concentration-dependent vasorelaxation; VRX0621688 was the most potent vasodilator. The KCNQ inhibitor XE991 (30 micromol/l) blocked the effects of both the drugs and ADRF. Inhibitors of cystathionine gamma lyase (CSE) beta-cyano-L-alanine (BCA, 5 mmol/l) and 4-propargyl glycine (PPG, 10 mmol/l) also blocked the relaxations. CSE is expressed in perivascular adipose tissue and endogenously generates H2S. The H2S donor NaHS produced concentration-dependent vasorelaxation, which was also blocked by XE991. The vasodilatory capacities of retigabine, VRX0530727, VRX0621238, and VRX0621688 were preserved following inhibition of H2S generation in perivascular fat. CONCLUSION We suggest that KCNQ channel opening is a powerful mechanism to produce vasorelaxation of systemic arteries in rats and mice. Furthermore, KCNQ channels play a major role in the paracrine control of vascular tone by perivascular adipose tissue, which is at least in part mediated or modulated by H2S. In conditions of reduced H2S release from perivascular adipose tissue, these paracrine effects can be mimicked by synthetic KCNQ channel openers.
Collapse
|
29
|
Abstract
Flupirtine is a centrally acting, non-opioid analgesic that is available in a number of European countries for the treatment of a variety of pain states. The therapeutic benefits seen with flupirtine relate to its unique pharmacological properties. Flupirtine displays indirect NDMA receptor antagonism via activation of potassium channels and is the first representative of a pharmacological class denoted the 'selective neuronal potassium channel openers'. The generation of the M-current is facilitated by flupirtine via the opening of neuronal Kv7 potassium channels. The opening of these channels inhibits exaggerated neuronal action potential generation and controls neuronal excitability. Neuronal hyperexcitability is a physiological component of many pain states such as chronic pain, migraine and neurogenic pain. Although large-scale clinical trials are lacking, the clinical trial database available to date from smaller-scale studies, together with extensive clinical experience, indicate that flupirtine effectively reduces chronic musculoskeletal pain, migraine and neuralgias, amongst other types of pain. In addition, flupirtine produces, at recommended clinical doses, muscle-relaxing effects in the presence of abnormally increased muscle tension. Its analgesic and muscle-relaxant properties were comparable to tramadol and chlormezanone, respectively, in two prospective trials in patients with lower back pain. Cytoprotective, anti-apoptotic and antioxidant properties have also been associated with flupirtine use in a small number of studies to date. When provided as combination therapy with morphine, flupirtine increases the antinociceptive activity of morphine 4-fold. Flupirtine displays superior tolerability when compared with tramadol and pentazocine. The most common adverse effects associated with flupirtine use are drowsiness, dizziness, heartburn, dry mouth, fatigue and nausea. With respect to its molecular structure, mechanism of action and adverse event profile, flupirtine is a unique drug. Flupirtine is an analgesic with many potential therapeutic benefits that may prove useful in the treatment of many disease states.
Collapse
Affiliation(s)
- Jacques Devulder
- Department of Anaesthesia, Section Pain Clinic, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
30
|
Sittl R, Carr RW, Schwarz JR, Grafe P. The Kv7 potassium channel activator flupirtine affects clinical excitability parameters of myelinated axons in isolated rat sural nerve. J Peripher Nerv Syst 2010; 15:63-72. [PMID: 20433607 DOI: 10.1111/j.1529-8027.2010.00253.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Flupirtine is an activator of Kv7 (KCNQ/M) potassium channels that has found clinical use as an analgesic with muscle relaxant properties. Kv7 potassium channels are expressed in axonal membranes and pharmacological activation of these channels may restore abnormal nerve excitability. We have examined the effect of flupirtine on the electrical excitability of myelinated axons in isolated segments of rat sural nerve. Axonal excitability was studied in vitro with the same parameters used by clinical neurophysiologists to assess peripheral nerve excitability in situ. Application of flupirtine in low micromolar concentrations resulted in an increase in threshold current, a reduction of refractoriness and an increase in post-spike superexcitability. These effects are consistent with an increase in Kv7 conductance and membrane hyperpolarization. Flupirtine also enhanced and prolonged the late, long-lasting period of axonal subexcitability that follows a short burst of action potentials. This effect was blocked by XE 991 (10 microM), an antagonist of Kv7 channels. In summary, flupirtine affects measures of excitability that are altered in the myelinated axons of patients with peripheral nerve disorders. This indicates that neuropathies with abnormal nerve excitability parameters corresponding to those affected by flupirtine may benefit from activation of axonal Kv7 potassium channels.
Collapse
Affiliation(s)
- Ruth Sittl
- Institute of Physiology, University of Munich, Munich, Germany.
| | | | | | | |
Collapse
|
31
|
Klawe C, Maschke M. Flupirtine: pharmacology and clinical applications of a nonopioid analgesic and potentially neuroprotective compound. Expert Opin Pharmacother 2009; 10:1495-500. [DOI: 10.1517/14656560902988528] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
32
|
Methling K, Reszka P, Lalk M, Vrana O, Scheuch E, Siegmund W, Terhaag B, Bednarski PJ. Investigation of the in vitro metabolism of the analgesic flupirtine. Drug Metab Dispos 2008; 37:479-93. [PMID: 19074524 DOI: 10.1124/dmd.108.024364] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The in vitro metabolism of flupirtine, ethyl-N-[2-amino-6-(4-fluorophenylmethyl-amino)pyridine-3-yl]carbamate, a centrally acting analgesic with muscle tone-reducing activity, was studied. Two flupirtine metabolites were already known: the N-acetylated analog D13223 and 4-fluorohippuric acid. The structure of flupirtine suggested that redox chemistry may play a role in metabolism, and cyclic voltammetry studies showed that the drug undergoes facile and irreversible redox reactions. Thus, oxidative metabolism was investigated first. With CYP3A1-induced rat liver microsomes an 18% turnover of flupirtine and a 20 to 25% turnover of D13223 took place over 30 min, but less than 5% turnover of flupirtine was observed with all human liver microsomal preparations tested, evidence that cytochrome P450 does not contribute appreciably to the metabolism in humans. Likewise, no involvement of human monoamine oxidase (isoforms A and B) was found for either flupirtine or D13223. In contrast, flupirtine was an excellent substrate for both human myeloperoxidase and horse radish peroxidase (HRP). These enzymes produced detectable amounts of oxidation products. Incubations of flupirtine with HRP produced an oxidation product that could be trapped with glutathione, the resulting glutathione conjugate was characterized by mass spectrometry and NMR. Metabolism of D13223 by both peroxidases was also observed but to a much lesser extent. Porcine liver esterases cleave the carbamate group of flupirtine, and both human N-acetyltransferases 1 and 2 acetylated the hydrolysis product, presumably descarboethoxyflupirtine, with nearly equal efficiencies to yield D13223. Incubations of human liver microsomes with flupirtine or the metabolite D13223 together with UDP-glucuronic acid gave two isomeric N-glucuronides in both cases.
Collapse
Affiliation(s)
- Karen Methling
- Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sättler MB, Williams SK, Neusch C, Otto M, Pehlke JR, Bähr M, Diem R. Flupirtine as neuroprotective add-on therapy in autoimmune optic neuritis. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1496-507. [PMID: 18832577 DOI: 10.2353/ajpath.2008.080491] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a common inflammatory disease of the central nervous system that results in persistent impairment in young adults. During chronic progressive disease stages, there is a strong correlation between neurodegeneration and disability. Current therapies fail to prevent progression of neurological impairment during these disease stages. Flupirtine, a drug approved for oral use in patients suffering from chronic pain, was used in a rat model of autoimmune optic neuritis and significantly increased the survival of retinal ganglion cells, the neurons that form the axons of the optic nerve. When flupirtine was combined with interferon-beta, an established immunomodulatory therapy for MS, visual functions of the animals were improved during the acute phase of optic neuritis. Furthermore, flupirtine protected retinal ganglion cells from degeneration in a noninflammatory animal model of optic nerve transection. Although flupirtine was shown previously to increase neuronal survival by Bcl-2 up-regulation, this mechanism does not appear to play a role in flupirtine-mediated protection of retinal ganglion cells either in vitro or in vivo. Instead, we showed through patch-clamp investigations that the activation of inwardly rectifying potassium channels is involved in flupirtine-mediated neuroprotection. Considering the few side effects reported in patients who receive long-term flupirtine treatment for chronic pain, our results indicate that this drug is an interesting candidate for further evaluation of its neuroprotective potential in MS.
Collapse
Affiliation(s)
- Muriel B Sättler
- Department of Neurology, University of Göttingen, Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
34
|
G-protein inwardly rectifying potassium channels are involved in the hypotensive effect of I1-imidazoline receptor selective ligands. J Hypertens 2008; 26:1025-32. [PMID: 18398346 DOI: 10.1097/hjh.0b013e3282f5ed44] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study examined the role of G-protein inwardly rectifying potassium (GIRK) channels in the depressor responses elicited by intracisternal injections of imidazoline-like drugs in anesthetized rabbits. METHODS AND RESULTS Intracisternal injections of the I1-imidazoline receptor (I1R) selective ligands LNP509 (30 microg/kg) and LNP640 (2 microg/kg) (subthreshold doses), and of the GIRK channel opener flupirtine (30 microg/kg) did not affect mean arterial blood pressure (MAP). LNP509 and LNP640, however, elicited substantial depressor responses in rabbits pretreated with flupirtine (-17 +/- 2 and -18 +/- 1 mmHg, respectively, P < 0.05). Injection of higher doses of LNP509 (200 microg/kg) or LNP640 (10 microg/kg) elicited substantial reductions in MAP (-45 +/- 3 and -39 +/- 2 mmHg, respectively, P < 0.05) in naive rabbits. The depressor responses elicited by the higher doses of LNP509 or LNP640 were markedly diminished by pretreatment with the GIRK channel blocker tertiapin-Q (10 microg/kg) (-23 +/- 3 and -26 +/- 2 mmHg, respectively, P < 0.05 compared with nonpretreated rabbits), whereas tertiapin-Q (10 microg/kg) did not affect MAP by itself. Maximal-specific binding (Bmax) of the I1R ligand [I]LNP911 to PC12 cell membranes (296 +/- 59 fmol/mg protein) was enhanced by flupirtine pretreatment whereas it was reduced by tertiapin-Q pretreatment (687 +/- 122 and 68 +/- 21 fmol/mg protein, respectively, P < 0.05 vs. control binding). CONCLUSION These findings demonstrate that the modulation of GIRK channels affects I1R's function and raise the possibility that GIRK channels, and I1Rs are parts of a single proteic complex.
Collapse
|
35
|
Lang F, Gulbins E, Szabo I, Vereninov A, Huber SM. Ion Channels, Cell Volume, Cell Proliferation and Apoptotic Cell Death. SENSING WITH ION CHANNELS 2008. [DOI: 10.1007/978-3-540-72739-2_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
Fawcett RJ, Osborne NN. Flupirtine attenuates sodium nitroprusside-induced damage to retinal photoreceptors, in situ. Brain Res Bull 2007; 73:278-88. [PMID: 17562394 DOI: 10.1016/j.brainresbull.2007.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/16/2007] [Accepted: 04/17/2007] [Indexed: 12/17/2022]
Abstract
Flupirtine has been shown to function as a neuroprotectant and is presently used in man to treat a number of conditions. The aim of this study was to investigate the specific antioxidant properties of flupirtine in relation to oxidant-induced damage to retinal photoreceptors. Initial in vitro studies on brain membranes showed that flupirtine was approximately 20 times more potent than trolox (vitamin E analogue) and 8 times more potent than metipranolol at attenuating lipid peroxidation caused by the nitric oxide donor, sodium nitroprusside (SNP). Subsequent immunohistochemical studies revealed that following an intraocular injection of SNP, retinal photoreceptors are the only retinal cell types that appear to be clearly affected. This was supported by electroretinogram (ERG) recordings which showed both the a- and b-wave amplitudes to be significantly reduced. Western blotting techniques showed that SNP caused a significant decrease in photoreceptor-specific markers (RET-P1, rhodopsin kinase), an increase in cleaved caspase-3, Bcl-2, and cleaved PARP proteins that are associated with apoptosis and no change in the ganglion cell specific marker, neurofilament (NF-L). This was supported by RT-PCR data where rhodopsin (photoreceptor specific) mRNA was reduced while Thy-1 and NF-L (ganglion cell specific) mRNAs were unaffected. In addition SNP caused an elevation of glial cell response mRNAs primarily associated with Müller cells (GFAP, CNTF, bFGF) as well as caspase-3 and Bcl-2. Importantly, when flupirtine was co-injected, the effects to the retina caused by SNP on retinal proteins and mRNAs were in most cases significantly blunted. The conclusion reached from this study is that flupirtine is a powerful antioxidant and when injected into the eye with SNP attenuates the detrimental influence of SNP to retinal photoreceptors. Since oxidative stress has been implicated in retinal diseases like age-related macular degeneration (AMD) this study provides "proof of principle" for the idea that flupirtine may help individuals suffering from such retinal diseases.
Collapse
Affiliation(s)
- R J Fawcett
- Nuffield Laboratory of Ophthalmology, Oxford University, Walton Street, Oxford OX2 6AW, UK
| | | |
Collapse
|
37
|
Lang F, Huber SM, Szabo I, Gulbins E. Plasma membrane ion channels in suicidal cell death. Arch Biochem Biophys 2007; 462:189-94. [PMID: 17316548 DOI: 10.1016/j.abb.2006.12.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 12/21/2006] [Accepted: 12/28/2006] [Indexed: 02/08/2023]
Abstract
The machinery leading to apoptosis includes altered activity of ion channels. The channels contribute to apoptotic cell shrinkage and modify intracellular ion composition. Cl(-) channels allow the exit of Cl(-), osmolytes and HCO(3)(-) leading to cell shrinkage and cytosolic acidification. K(+) exit through K(+) channels contributes to cell shrinkage and decreases intracellular K(+) concentration, which in turn favours apoptotic cell death. K(+) channel activity further determines the cell membrane potential, a driving force for Ca(2+) entry through Ca(2+) channels. Ca(2+) may enter through unselective cation channels. An increase of cytosolic Ca(2+) may stimulate several enzymes executing apoptosis. Specific ion channel blockers may either promote or counteract suicidal cell death. The present brief review addresses the role of ion channels in the regulation of suicidal cell death with special emphasis on the role of channels in CD95 induced apoptosis of lymphocytes and suicidal death of erythrocytes or eryptosis.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tübingen, Gmelinstrasse 5, D72076 Tuebingen, Germany.
| | | | | | | |
Collapse
|
38
|
Lang F, Föller M, Lang K, Lang P, Ritter M, Vereninov A, Szabo I, Huber SM, Gulbins E. Cell volume regulatory ion channels in cell proliferation and cell death. Methods Enzymol 2007; 428:209-25. [PMID: 17875419 DOI: 10.1016/s0076-6879(07)28011-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alterations of cell volume are key events during both cell proliferation and apoptotic cell death. Cell proliferation eventually requires an increase of cell volume, and apoptosis is typically paralleled by cell shrinkage. Alterations of cell volume require the participation of ion transport across the cell membrane, including appropriate activity of Cl(-) and K(+) channels. Cl(-) channels modify cytosolic Cl(-) activity and mediate osmolyte flux, and thus influence cell volume. Most Cl(-) channels allow exit of HCO(3)(-), leading to cytosolic acidification, which in turn inhibits cell proliferation and favors apoptosis. K(+) exit through K(+) channels decreases cytosolic K(+) concentration, which may sensitize the cell for apoptotic cell death. K(+) channel activity further maintains the cell membrane potential, a critical determinant of Ca(2+) entry through Ca(2+) channels. Ca(2+) may, in addition, enter through Ca(2+)-permeable cation channels, which, in some cells, are activated by hyperosmotic shock. Increases of cytosolic Ca(2+) activity may trigger both mechanisms required for cell proliferation and mechanisms, leading to apoptosis. Thereby cell proliferation and apoptosis depend on magnitude and temporal organization of Ca(2+) entry, as well as activity of other signaling pathways. Accordingly, the same ion channels may participate in the stimulation of both cell proliferation and apoptosis. Specific ion channel blockers may thus abrogate both cellular mechanisms, depending on cell type and condition.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Binzen U, Greffrath W, Hennessy S, Bausen M, Saaler-Reinhardt S, Treede RD. Co-expression of the voltage-gated potassium channel Kv1.4 with transient receptor potential channels (TRPV1 and TRPV2) and the cannabinoid receptor CB1 in rat dorsal root ganglion neurons. Neuroscience 2006; 142:527-39. [PMID: 16889902 DOI: 10.1016/j.neuroscience.2006.06.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 05/22/2006] [Accepted: 06/09/2006] [Indexed: 01/20/2023]
Abstract
Potassium channels contribute to basic neuronal excitability and modulation. Here, we examined expression patterns of the voltage-gated potassium channel Kv1.4, the nociceptive transduction channels TRPV1 and TRPV2 as well as the putative anti-nociceptive cannabinoid receptor CB1 by immunofluorescence double-labelings in sections of rat dorsal root ganglia (DRGs). Kv1.4, TRPV1 and CB1 were each detected in about one third of neurons (35.7+/-0.5%, 29.4+/-1.1% and 36.4+/-0.5%, respectively, mean diameter 19.1+/-0.3 microm). TRPV2 was present in 4.4+/-0.4% of all neurons that were significantly larger in diameter (27.4+/-0.7 microm; P < 0.001). Antibody double-labeling revealed that the majority of Kv1.4-positive neurons co-expressed TRPV1 (73.9+/-1.5%) whereas none expressed TRPV2. The largest overlap was found with CB1 (93.1+/-0.1%). CB1 expression resembled that seen for Kv1.4 since the majority of neurons expressing CB1-protein also expressed TRPV1 (69.4+/-6.5%) but not TRPV2 (0.6+/-0.3%). When CB1-mRNA was detected using in situ hybridizations an additional subset of larger neurons was labeled including 82.4+/-17.7% of the TRPV2 expressing neurons. However, co-localization of Kv1.4 with CB1-mRNA (92%, mean diameter: 18.5 microm) was essentially the same as with CB1-protein. The almost complete overlap of CB1 and Kv1.4 in nociceptive DRG neurons suggests a functional synergistic action between Kv1.4 and CB1. The potassium channel may have two important roles in nociception. As the molecular basis of A-type current it could be involved in the control of repetitive discharges at peripheral terminals and as a downstream signal transduction site of CB1 in the control of presynaptic transmitter release at central terminals.
Collapse
Affiliation(s)
- U Binzen
- Institute of Physiology and Pathophysiology, Johannes Gutenberg-University, Saarstrasse 21, D-55099, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Lang F, Föller M, Lang KS, Lang PA, Ritter M, Gulbins E, Vereninov A, Huber SM. Ion channels in cell proliferation and apoptotic cell death. J Membr Biol 2006; 205:147-57. [PMID: 16362503 DOI: 10.1007/s00232-005-0780-5] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Indexed: 12/11/2022]
Abstract
Cell proliferation and apoptosis are paralleled by altered regulation of ion channels that play an active part in the signaling of those fundamental cellular mechanisms. Cell proliferation must--at some time point--increase cell volume and apoptosis is typically paralleled by cell shrinkage. Cell volume changes require the participation of ion transport across the cell membrane, including appropriate activity of Cl- and K+ channels. Besides regulating cytosolic Cl- activity, osmolyte flux and, thus, cell volume, most Cl- channels allow HCO3- exit and cytosolic acidification, which inhibits cell proliferation and favors apoptosis. K+ exit through K+ channels may decrease intracellular K+ concentration, which in turn favors apoptotic cell death. K+ channel activity further maintains the cell membrane potential, a critical determinant of Ca2+ entry through Ca2+ channels. Cytosolic Ca2+ may trigger mechanisms required for cell proliferation and stimulate enzymes executing apoptosis. The switch between cell proliferation and apoptosis apparently depends on the magnitude and temporal organization of Ca2+ entry and on the functional state of the cell. Due to complex interaction with other signaling pathways, a given ion channel may play a dual role in both cell proliferation and apoptosis. Thus, specific ion channel blockers may abrogate both fundamental cellular mechanisms, depending on cell type, regulatory environment and condition of the cell. Clearly, considerable further experimental effort is required to fully understand the complex interplay between ion channels, cell proliferation and apoptosis.
Collapse
Affiliation(s)
- F Lang
- Department of Physiology, University of Tübingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Föller M, Kasinathan RS, Duranton C, Wieder T, Huber SM, Lang F. PGE 2-induced Apoptotic Cell Death in K562 Human Leukaemia Cells. Cell Physiol Biochem 2006; 17:201-10. [PMID: 16790996 DOI: 10.1159/000094125] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Prostaglandin-E2 (PGE2) is known to trigger suicidal death of nucleated cells (apoptosis) and enucleated erythrocytes (eryptosis). In erythrocytes PGE2 induced suicidal cell death involves activation of nonselective cation channels leading to Ca2+ entry followed by cell shrinkage and triggering of Ca2+ sensitive cell membrane scrambling with phosphatidylserine (PS) exposure at the cell surface. The present study was performed to explore whether PGE2 induces apoptosis of nucleated cells similarly through cation channel activation and to possibly disclose the molecular identity of the cation channels involved. To this end, Ca2+ activity was estimated from Fluo3 fluorescence, mitochondrial potential from DePsipher fluorescence, phosphatidylserine exposure from annexin binding, caspase activation from caspAce fluorescence, cell volume from FACS forward scatter, and DNA fragmentation utilizing a photometric enzyme immunoassay. Stimulation of K562 human leukaemia cells with PGE2 (50 microM) increased cytosolic Ca2+ activity, decreased forward scatter, depolarized the mitochondrial potential, increased annexin binding, led to caspase activation and resulted in DNA fragmentation. Gene silencing of the Ca2+-permeable transient receptor potential cation channel TRPC7 significantly blunted PGE2-induced triggering of PS exposure and DNA fragmentation. In conclusion, K562 cells express Ca2+-permeable TRPC7 channels, which are activated by PGE2 and participate in the triggering of apoptosis.
Collapse
Affiliation(s)
- Michael Föller
- Department of Physiology, University of Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Wickenden AD, Roeloffs R, McNaughton-Smith G, Rigdon GC. KCNQ potassium channels: drug targets for the treatment of epilepsy and pain. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.14.4.457] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Kobayashi T, Washiyama K, Ikeda K. Modulators of G protein-activated inwardly rectifying K+ channels: potentially therapeutic agents for addictive drug users. Ann N Y Acad Sci 2005; 1025:590-4. [PMID: 15542767 DOI: 10.1196/annals.1316.073] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
G protein-activated inwardly rectifying K+ (GIRK, Kir3) channels play an important role in the inhibitory regulation of neuronal excitability in most brain regions and heart rate through activation of various G protein-coupled receptors, such as opioid, cannabinoid, and D2 dopamine receptors. Therefore, modulators of GIRK channels may affect many brain functions. We have shown using Xenopus oocyte expression assays that ethanol directly activates GIRK channels, whereas various antipsychotics (thioridazine, clozapine, pimozide, and haloperidol) inhibit the channels. Here we investigated not only the effects of various selective serotonin reuptake inhibitor (SSRI) antidepressants (fluoxetine, citalopram, fluvoxamine, and zimelidine) and risperidone, an atypical antipsychotic, on GIRK channels, but also those of the various drugs tested on other Kir channels using the Xenopus oocyte system. Fluoxetine inhibited GIRK channels, whereas the other SSRIs and risperidone had a small or no effect on the channels. In contrast, Kir1.1 and Kir2.1 channels were insensitive to ethanol and various SSRIs and antipsychotics, although thioridazine weakly inhibited Kir1.1 channels. It has been shown that the function of GIRK channels is involved in seizure susceptibility, antinociception by opioids, cannabinoids, or ethanol, and cocaine reinforcement in studies using GIRK knockout mice and weaver mutant mice that have mutant GIRK2 channels insensitive to G proteins and ethanol. Activation of GIRK channels by opioids, cannabinoids, or ethanol may be one of these key effects. Therefore, GIRK channel modulators might be potential agents for the treatment of users of addictive drugs, such as cocaine, opioids, cannabinoids, and ethanol, as well as for the treatment of epilepsy and pain.
Collapse
Affiliation(s)
- Toru Kobayashi
- Department of Molecular Neuropathology, Brain Research Institute, Niigata University, Niigata, Niigata 951-8585, Japan.
| | | | | |
Collapse
|
44
|
Azad SC, Eder M, Simon W, Hapfelmeier G, Dodt HU, Zieglgänsberger W, Rammes G. The potassium channel modulator flupirtine shifts the frequency–response function of hippocampal synapses to favour LTD in mice. Neurosci Lett 2004; 370:186-90. [PMID: 15488320 DOI: 10.1016/j.neulet.2004.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2004] [Revised: 08/02/2004] [Accepted: 08/10/2004] [Indexed: 11/26/2022]
Abstract
Flupirtine is a centrally acting nonopioid analgesic with muscle-relaxant properties. Flupirtine has been found to activate inwardly rectifying potassium conductances and hence to indirectly inhibit the activation of NMDA receptors. NMDA receptor activation is crucial for the induction of long-term potentiation (LTP) of synaptic transmission, which is considered as cellular correlate of learning and memory and of central sensitization in chronic pain states. Although flupirtine has been widely used for the management of pain, its effects on synaptic plasticity have not yet been investigated. We, therefore, performed extracellular and whole-cell patch-clamp recordings in hippocampal slices of mice to examine the effects of flupirtine on synaptic plasticity and neuronal membrane properties. Excitatory postsynaptic potentials (EPSPs) in the CA1 region were evoked alternately by stimulating two independent Schaffer collateral-commissural inputs. LTP and long-term depression (LTD) were induced by different stimulation paradigms (100 Hz, 10 Hz, 5 Hz, and 1 Hz). Flupirtine (30 microM) diminished the degree of LTP and enhanced LTD. This effect is most likely due to the hyperpolarization of CA1 pyramidal neurons and the reduction of their input resistance found after application of flupirtine. The observed effects on synaptic strength could underly the beneficial effects of flupirtine on different types of chronic pain.
Collapse
Affiliation(s)
- Shahnaz Christina Azad
- Max-Planck-Institute of Psychiatry, Clinical Neuropharmacology, Kraepelinstrasse 2, 80804 München, Germany.
| | | | | | | | | | | | | |
Collapse
|
45
|
Do Y, McKallip RJ, Nagarkatti M, Nagarkatti PS. Activation through Cannabinoid Receptors 1 and 2 on Dendritic Cells Triggers NF-κB-Dependent Apoptosis: Novel Role for Endogenous and Exogenous Cannabinoids in Immunoregulation. THE JOURNAL OF IMMUNOLOGY 2004; 173:2373-82. [PMID: 15294950 DOI: 10.4049/jimmunol.173.4.2373] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The precise role of cannabinoid receptors (CB)1 and CB2, as well as endogenous ligands for these receptors, on immune cells remains unclear. In the current study, we examined the effect of endogenous and exogenous cannabinoids on murine bone marrow-derived dendritic cells (DCs). Addition of Delta(9)-tetrahydrocannabinol (THC), a major psychoactive component found in marijuana or anandamide, an endogenous cannabinoid, to DC cultures induced apoptosis in DCs. DCs expressed CB1 and CB2 receptors and the engagement of both receptors was necessary to trigger apoptosis. Treatment with THC induced caspase-2, -8, and -9 activation, cleavage of Bid, decreased mitochondrial membrane potential, and cytochrome c release, suggesting involvement of death-receptor and mitochondrial pathways. DCs from Bid-knockout mice were sensitive to THC-induced apoptosis thereby suggesting that Bid was dispensable. There was no induction of p44/p42 MAPK, p38 MAPK, or stress-activated protein/JNK pathway in THC-treated DCs. However, THC treatment induced phosphorylation of IkappaB-alpha, and enhanced the transcription of several apoptotic genes regulated by NF-kappaB. Moreover, inhibition of NF-kappaB was able to block THC-induced apoptosis in DCs. Lastly, in vivo treatment of mice with THC caused depletion of splenic DCs. Together, our study demonstrates for the first time that endogenous and exogenous cannabinoids may suppress the immune response through their ability to induce apoptosis in DCs.
Collapse
Affiliation(s)
- Yoonkyung Do
- Department of Microbiology and Immunology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
46
|
Downer EJ, Fogarty MP, Campbell VA. Tetrahydrocannabinol-induced neurotoxicity depends on CB1 receptor-mediated c-Jun N-terminal kinase activation in cultured cortical neurons. Br J Pharmacol 2004; 140:547-57. [PMID: 14522843 PMCID: PMC1574055 DOI: 10.1038/sj.bjp.0705464] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Delta9-Tetrahydrocannabinol (THC), the main psychoactive ingredient of marijuana, induces apoptosis in cultured cortical neurons. THC exerts its apoptotic effects in cortical neurons by binding to the CB1 cannabinoid receptor. The CB1 receptor has been shown to couple to the stress-activated protein kinase, c-Jun N-terminal kinase (JNK). However, the involvement of specific JNK isoforms in the neurotoxic properties of THC remains to be established. The present study involved treatment of rat cultured cortical neurons with THC (0.005-50 microM), and combinations of THC with the CB1 receptor antagonist, AM 251 (10 microM) and pertussis toxin (PTX; 200 ng ml-1). Antisense oligonucleotides (AS) were used to deplete neurons of JNK1 and JNK2 in order to elucidate their respective roles in THC signalling. Here we report that THC induces the activation of JNK via the CB1 receptor and its associated G-protein, Gi/o. Treatment of cultured cortical neurons with THC resulted in a differential timeframe of activation of the JNK1 and JNK2 isoforms. Use of specific JNK1 and JNK2 AS identified activation of caspase-3 and DNA fragmentation as downstream consequences of JNK1 and JNK2 activation. The results from this study demonstrate that activation of the CB1 receptor induces JNK and caspase-3 activation, an increase in Bax expression and DNA fragmentation. The data demonstrate that the activation of both JNK1 and JNK2 isoforms is central to the THC-induced activation of the apoptotic pathway in cortical neurons.
Collapse
Affiliation(s)
- Eric J Downer
- Department of Physiology, Trinity College, Trinity College Institute of Neuroscience, Dublin 2, Ireland
| | - Marie P Fogarty
- Department of Physiology, Trinity College, Trinity College Institute of Neuroscience, Dublin 2, Ireland
| | - Veronica A Campbell
- Department of Physiology, Trinity College, Trinity College Institute of Neuroscience, Dublin 2, Ireland
- Author for correspondence:
| |
Collapse
|
47
|
Hoffmann O, Gommert LR, Egert M. Paradoxical Cerebral Cortical Hyperexcitability Following Flupirtine Overdose. ACTA ACUST UNITED AC 2004; 42:913-6. [PMID: 15533031 DOI: 10.1081/clt-200035096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We report the case of a patient with increased cerebral cortical excitability following intoxication with flupirtine, a centrally acting analgesic and antispastic drug. Typically, flupirtine exerts membrane stabilizing and hyperpolarizing effects through activation of neuronal G-protein regulated inwardly rectifying potassium channels (GIRK). Based on these properties, GIRK activators have been suggested as candidates for antiepileptic drug development. In contrast, our observation suggests that these substances may also display unexpected proconvulsant effects in vivo.
Collapse
Affiliation(s)
- Olaf Hoffmann
- Department of Neurology, Charité-Hochschulmedizin, Berlin, Germany.
| | | | | |
Collapse
|
48
|
Ford JW, Stevens EB, Treherne JM, Packer J, Bushfield M. Potassium channels: gene family, therapeutic relevance, high-throughput screening technologies and drug discovery. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2002; 58:133-68. [PMID: 12079199 DOI: 10.1007/978-3-0348-8183-8_4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Existing drugs that modulate ion channels represent a key class of pharmaceutical agents across many therapeutic areas and there is considerable further potential for potassium channel drug discovery. Potassium channels represent the largest and most diverse sub-group of ion channels and they play a central role in regulating the membrane potential of cells. Recent advances in genomics have greatly added to the number of these potential drug targets, but selecting a suitable potassium channel for drug discovery research is a key step. In particular, the potential therapeutic relevance of a potassium channel should be taken into account when selecting a target for screening. Potassium channel drug discovery is being driven by a need to identify lead compounds that can provide tractable starting points for medicinal chemistry. Furthermore, advances in laboratory automation have brought significant opportunities to increase screening throughput for potassium channel assays, but careful assay configuration to model drug-target interactions in a physiological manner is an essential consideration. Several potassium channel screening platforms are described in this review in order to provide some insight into the variety of formats available for screening, together with some of their inherent advantages and limitations. Particular emphasis is placed on the mechanistic basis of drug-target interaction and those aspects of structure/function that are of prime importance in potassium channel drug discovery.
Collapse
|
49
|
Campbell VA. Tetrahydrocannabinol-induced apoptosis of cultured cortical neurones is associated with cytochrome c release and caspase-3 activation. Neuropharmacology 2001; 40:702-9. [PMID: 11311898 DOI: 10.1016/s0028-3908(00)00210-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Delta(9)-tetrahydrocannabinol (THC), the principal psychoactive component of marijuana, is associated with impaired cognition and altered cortical function. THC transduces its central effects via activation of the G-protein linked cannabinoid receptor CB1. In this study we report that THC induces morphological degenerative changes in cultured cortical neurones, such as membrane blebbing and formation of apoptotic bodies, that are consistent with the apoptotic pathway of cell death. The THC-induced apoptosis was blocked by the CB1 receptor antagonist AM251 and pertussis toxin (PTX), suggesting that this effect of THC involves receptor-mediated activation of the G-protein subtypes G(i) or G(o). THC also promoted translocation of mitochondrial cytochrome c to the cytosol and increased the activity of the cysteine protease caspase-3, in a PTX-sensitive manner. The results from this study suggest that coupling of THC to a PTX-sensitive G-protein promotes cytochrome c release, caspase-3 activation and subsequent degeneration of cultured cortical neurones. This apoptotic pathway may underlie the compromised neuronal function that is associated with marijuana usage.
Collapse
Affiliation(s)
- V A Campbell
- Department of Physiology, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
50
|
Lawson K. Is there a role for potassium channel openers in neuronal ion channel disorders? Expert Opin Investig Drugs 2000; 9:2269-80. [PMID: 11060806 DOI: 10.1517/13543784.9.10.2269] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malfunction in ion channels, due to mutations in genes encoding channel proteins or the presence of autoantibodies, are increasing being implicated in causing disease conditions, termed channelopathies. Dysfunction of potassium (K(+)) channels has been associated with the pathophysiology of a number of neurological, as well as peripheral, disorders (e.g., episodic ataxia, epilepsy, neuromyotonia, Parkinson's disease, congenital deafness, long QT syndrome). K(+) channels, which demonstrate a high degree of diversity and ubiquity, are fundamental in the control of membrane depolarisation and cell excitability. A common feature of K(+) channelopathies is a reduction or loss of membrane potential repolarisation. The identification of K(+) channel subtype specific openers will allow the recovery of the mechanism(s) responsible for counteraction of uncontrolled cellular depolarisation. Synthetic agents that demonstrate K(+) channel opening properties are available for a variety of K(+) channel subtypes (e.g., K(ATP), BK(Ca), GIRK and M-channel). This study reviews the realistic therapeutic potential that may be gained in a broad spectrum of clinical conditions by K(+) channel openers. K(+) channel openers would therefore identify dysfunctional K(+) channel as therapeutic targets for clinical benefit, in addition being able to modulate normally functioning K(+) channels to gain clinical management of pathophysiological events irrespective of the cause.
Collapse
Affiliation(s)
- K Lawson
- Biomedical Research Centre, Sheffield Hallam University, School of Science and Mathematics, City Campus, Sheffield, S1 1WB, UK.
| |
Collapse
|