1
|
Ilepcimide inhibited sodium channel activity in mouse hippocampal neurons. Epilepsy Res 2020; 170:106533. [PMID: 33385943 DOI: 10.1016/j.eplepsyres.2020.106533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/29/2020] [Accepted: 12/07/2020] [Indexed: 11/23/2022]
Abstract
Ilepcimide (ICM), a clinically effective antiepileptic drug, has been used in China for decades; however, its antiepileptic mechanism remains unclear. ICM is structurally similar to antiepileptic drug lamotrigine (LTG). LTG exerts its anticonvulsant effect by inhibiting voltage-gated Na+ channel (NaV) activity. Thus it is speculated that ICM also exert its antiepileptic activity by inhibiting sodium channel activity. We studied the inhibition of NaV activity by ICM in acutely isolated mouse hippocampal pyramidal neurons. We evaluated ICM-mediated tonic, concentration-dependent, and voltage-dependent inhibition of NaV, and the effects of ICM and LTG on NaV biophysical properties. Na+ currents in hippocampal pyramidal neurons were tonically inhibited by ICM in a concentration- and voltage-dependent manner. The half-maximal inhibitory concentration (IC50) of ICM at a holding potential (Vh) of -90 mV was higher than that at a Vh of -70 mV. Compared with the control groups, in the presence of 10 μM ICM, the current densities of Na+ channels were reduced, the half-maximal availability of the inactivation curve (V1/2) was shifted to more negative potentials, and the recovery from inactivation was delayed. These data can contribute to further investigation of the inhibitory effect of ICM on the sodium channel, suggesting that the main reason for the anticonvulsant effect of ICM is the small influx of sodium ions. ICM can prevent abnormal discharge of neurons, which may prevent epilepsy.
Collapse
|
2
|
Peng YS, Wu HT, Lai YC, Chen JL, Yang YC, Kuo CC. Inhibition of neuronal Na+ currents by lacosamide: Differential binding affinity and kinetics to different inactivated states. Neuropharmacology 2020; 179:108266. [DOI: 10.1016/j.neuropharm.2020.108266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/06/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
|
3
|
Robles-Gómez E, Benítez-Villalobos F, Soriano-García M, Antúnez-Argüelles E. Non-peptide molecules in the pedicellariae of Toxopneustes roseus. Toxicon 2020; 184:143-151. [DOI: 10.1016/j.toxicon.2020.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 11/30/2022]
|
4
|
Lo YC, Kuo CC. Temperature Dependence of the Biophysical Mechanisms Underlying the Inhibition and Enhancement Effect of Amiodarone on hERG Channels. Mol Pharmacol 2019; 96:330-344. [PMID: 31253645 DOI: 10.1124/mol.119.116400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/21/2019] [Indexed: 11/22/2022] Open
Abstract
hERG K+ channel is important for controlling the duration of cardiac action potentials. Amiodarone (AMD), a widely prescribed class III antiarrhythmic, could inhibit hERG currents with relatively few tachyarrhythmic adverse events. We use injected Xenopus oocyte with two-electrode voltage clamp techniques to characterize the action of AMD on hERG channels. We found that AMD binds to the resting hERG channel with an apparent dissociation constant of ∼1.4 μM, and inhibits hERG currents at mild and strong depolarization pulses by slowing activation and enhancing inactivation, respectively, at 22°C. The activation kinetics of hERG channel activation are much faster, but inactivation kinetics are slower at 37°C. AMD accordingly has a 15% to 20% weaker and stronger inhibitory effect at mild and strong depolarization (e.g., -60 and +30 mV, 0.3-second pulse), respectively. In the meanwhile, the resurgent hERG tail currents are dose-dependently inhibited by AMD without altering the kinetics of current decay at both 22°C and 37°C, indicating facilitation of recovery from inactivation via the silent route. Most importantly, AMD no longer inhibits but enhances hERG currents at a mild pulse shortly after a prepulse at 37°C, but not so much at 22°C. We conclude that AMD is an effective hERG channel-gating modifier capable of lengthening the plateau phase of cardiac action potential (without increasing the chance of afterdepolarization). AMD, however, should be used with caution in hypothermia or the other scenarios that slow hERG channel activation. SIGNIFICANCE STATEMENT: It is known that amiodarone (AMD) acts on hERG K+ channels to treat cardiac arrhythmias with relatively little arrhythmogenicity. We found that AMD enhances hERG channel inactivation but slows activation as well as recovery from inactivation, and thus has a differential inhibition and enhancement effect on hERG currents at different phases of membrane voltage changes, especially at 37°C, but not so much at 22°C. AMD is therefore a relatively ideal agent against tachyarrhythmia at 37°C, but should be more cautiously used at lower temperatures or relevant pathophysiological/pharmacological scenarios associated with slower hERG channel activation because of the increased chances of adverse events.
Collapse
Affiliation(s)
- Yung-Chen Lo
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan; and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Chin Kuo
- Department of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan; and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
5
|
Turner AL, Perry MS. Outside the box: Medications worth considering when traditional antiepileptic drugs have failed. Seizure 2017; 50:173-185. [PMID: 28704741 DOI: 10.1016/j.seizure.2017.06.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Review and discuss medications efficacious for seizure control, despite primary indications for other diseases, as treatment options in patients who have failed therapy with traditional antiepileptic drugs (AEDs). METHODS Literature searches were conducted utilizing PubMed and MEDLINE databases employing combinations of search terms including, but not limited to, "epilepsy", "refractory", "seizure", and the following medications: acetazolamide, amantadine, bumetanide, imipramine, lidocaine, verapamil, and various stimulants. RESULTS Data from relevant case studies, retrospective reviews, and available clinical trials were gathered, analyzed, and reported. Experience with acetazolamide, amantadine, bumetanide, imipramine, lidocaine, verapamil, and various stimulants show promise for cases of refractory epilepsy in both adults and children. Many medications lack large scale, randomized clinical trials, but the available data is informative when choosing treatment for patients that have failed traditional epilepsy therapies. CONCLUSIONS All neurologists have encountered a patient that failed nearly every AED, diet, and surgical option. For these patients, we often seek fortuitous discoveries within small series and case reports, hoping to find a treatment that might help the patient. In the present review, we describe medications for which antiepileptic effect has been ascribed after they were introduced for other indications.
Collapse
Affiliation(s)
- Adrian L Turner
- Department of Pharmacy, Cook Children's Medical Center, 1500 Cooper Street, 4th Floor, Fort Worth, TX, 76104, USA
| | - M Scott Perry
- Comprehensive Epilepsy Program, Jane and John Justin Neurosciences Center, Cook Children's Medical Center, Fort Worth, TX, USA.
| |
Collapse
|
6
|
Föhr KJ, Knippschild U, Herkommer A, Fauler M, Peifer C, Georgieff M, Adolph O. State-dependent block of voltage-gated sodium channels by the casein-kinase 1 inhibitor IC261. Invest New Drugs 2017; 35:277-289. [PMID: 28164251 DOI: 10.1007/s10637-017-0429-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/12/2017] [Indexed: 12/13/2022]
Abstract
Background and Purpose IC261 (3-[(2,4,6-trimethoxyphenyl)methylidenyl]-indolin-2-one) has previously been introduced as an isoform specific inhibitor of casein kinase 1 (CK1) causing cell cycle arrest or cell death of established tumor cell lines. However, it is reasonable to assume that not all antitumor activities of IC261 are mediated by the inhibition of CK1. Meanwhile there is growing evidence that functional voltage-gated sodium channels are also implicated in the progression of tumors as their blockage suppresses tumor migration and invasion of different tumor cell lines. Thus, we asked whether IC261 functionally inhibits voltage-gated sodium channels. Experimental Approach Electrophysiological experiments were performed using the patch-clamp technique at human heart muscle sodium channels heterologously expressed in human TsA cells. Key Results IC261 inhibits sodium channels in a state-dependent manner. IC261 does not interact with the open channel and has only a low affinity for the resting state of the hNav1.5 (human voltage-gated sodium channel; Kr: 120 μM). The efficacy of IC261 strongly increases with membrane depolarisation, indicating that the inactivated state is an important target. The results of different experimental approaches finally revealed an affinity of IC261 to the inactivated state between 1 and 2 μM. Conclusion and Implications IC261 inhibits sodium channels at a similar concentration necessary to reduce CK1δ/ε activity by 50% (IC50 value 1 μM). Thus, inhibition of sodium channels might contribute to the antitumor activity of IC261.
Collapse
Affiliation(s)
- Karl J Föhr
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89075, Ulm, Germany.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Anna Herkommer
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, University of Ulm, Albert-Einstein-Allee 11, D-89081, Ulm, Germany
| | - Christian Peifer
- Institute of Pharmacy, University of Kiel, Gutenbergstr. 76, D-24118, Kiel, Germany
| | - Michael Georgieff
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89075, Ulm, Germany
| | - Oliver Adolph
- Department of Anesthesiology, University Hospital of Ulm, Albert-Einstein-Allee 23, D-89075, Ulm, Germany
| |
Collapse
|
7
|
Abstract
Voltage-gated sodium channels (VGSCs) are fundamentally important for the generation and coordinated transmission of action potentials throughout the nervous system. It is, therefore, unsurprising that they have been shown to play a central role in the genesis and alleviation of epilepsy. Genetic studies on patients with epilepsy have identified more than 700 mutations among the genes that encode for VGSCs attesting to their role in pathogenesis. Further, many common antiepileptic drugs act on VGSCs to suppress seizure activity. Here, we present an account of the role of VGSCs in epilepsy, both through their pathogenic dysfunction and as targets for pharmacotherapy.
Collapse
|
8
|
Nardi A, Damann N, Hertrampf T, Kless A. Advances in targeting voltage-gated sodium channels with small molecules. ChemMedChem 2012; 7:1712-40. [PMID: 22945552 DOI: 10.1002/cmdc.201200298] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/30/2012] [Indexed: 12/19/2022]
Abstract
Blockade of voltage-gated sodium channels (VGSCs) has been used successfully in the clinic to enable control of pathological firing patterns that occur in conditions as diverse as chronic pain, epilepsy, and arrhythmias. Herein we review the state of the art in marketed sodium channel inhibitors, including a brief compendium of their binding sites and of the cellular and molecular biology of sodium channels. Despite the preferential action of this drug class toward over-excited cells, which significantly limits potential undesired side effects on other cells, the need to develop a second generation of sodium channel inhibitors to overcome their critical clinical shortcomings is apparent. Current approaches in drug discovery to deliver novel and truly innovative sodium channel inhibitors is next presented by surveying the most recent medicinal chemistry breakthroughs in the field of small molecules and developments in automated patch-clamp platforms. Various strategies aimed at identifying small molecules that target either particular isoforms of sodium channels involved in specific diseases or anomalous sodium channel currents, irrespective of the isoform by which they have been generated, are critically discussed and revised.
Collapse
Affiliation(s)
- Antonio Nardi
- Global Drug Discovery, Department of Medicinal Chemistry, Grünenthal, Zieglerstrasse 6, 52078 Aachen, Germany.
| | | | | | | |
Collapse
|
9
|
Toledo E, Lebel A, Becerra L, Minster A, Linnman C, Maleki N, Dodick DW, Borsook D. The young brain and concussion: imaging as a biomarker for diagnosis and prognosis. Neurosci Biobehav Rev 2012; 36:1510-31. [PMID: 22476089 PMCID: PMC3372677 DOI: 10.1016/j.neubiorev.2012.03.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/15/2012] [Accepted: 03/21/2012] [Indexed: 01/20/2023]
Abstract
Concussion (mild traumatic brain injury (mTBI)) is a significant pediatric public health concern. Despite increased awareness, a comprehensive understanding of the acute and chronic effects of concussion on central nervous system structure and function remains incomplete. Here we review the definition, epidemiology, and sequelae of concussion within the developing brain, during childhood and adolescence, with current data derived from studies of pathophysiology and neuroimaging. These findings may contribute to a better understanding of the neurological consequences of traumatic brain injuries, which in turn, may lead to the development of brain biomarkers to improve identification, management and prognosis of pediatric patients suffering from concussion.
Collapse
Affiliation(s)
- Esteban Toledo
- Center for Pain and the Brain, Children's Hospital Boston, Harvard Medical School, United States
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Zhan XQ, He YL, Yao JJ, Zhuang JL, Mei YA. The antidepressant citalopram inhibits delayed rectifier outward K+ current in mouse cortical neurons. J Neurosci Res 2011; 90:324-36. [DOI: 10.1002/jnr.22744] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 06/08/2011] [Accepted: 06/21/2011] [Indexed: 01/04/2023]
|
11
|
Searching for novel anti-myotonic agents: pharmacophore requirement for use-dependent block of skeletal muscle sodium channels by N-benzylated cyclic derivatives of tocainide. Neuromuscul Disord 2011; 22:56-65. [PMID: 21802953 PMCID: PMC3314985 DOI: 10.1016/j.nmd.2011.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/20/2011] [Accepted: 07/04/2011] [Indexed: 11/22/2022]
Abstract
Drug screening on sodium currents of native myofibers by means of voltage-clamp recordings is predictive of pre-clinical anti-myotonic activity in vivo and ex vivo. By this approach we identified the N-benzylated beta-proline derivative of tocainide (To10) as the most potent use-dependent blocker of Nav1.4 so far. We tested novel analogs with modifications on the pharmacophore groups of To10. The substitution of the proline cycle with less planar piperidine or piperazine rings disclosed the importance of a two carbon atom distance and/or an additional nitrogen atom for potency. Structural changes on the xylididic group corroborated the role of a proper electronic cloud for hydrophobic interactions with the binding site. The N-benzylated moiety lead to a stereoselective behavior only in the rigid alpha-proline analog To11 vs. To10 and N-benzylated tocainide (To12). The results confirm the strict structural requirements of Nav1.4 blockers and allow to refine the drug design toward novel anti-myotonic drugs.
Collapse
|
12
|
Jo S, Bean BP. Inhibition of neuronal voltage-gated sodium channels by brilliant blue G. Mol Pharmacol 2011; 80:247-57. [PMID: 21536754 DOI: 10.1124/mol.110.070276] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Brilliant blue G (BBG), best known as an antagonist of P2X7 receptors, was found to inhibit voltage-gated sodium currents in N1E-115 neuroblastoma cells. Sodium currents elicited from a holding potential of -60 mV were blocked with an IC(50) of 2 μM. Block was enhanced in a use-dependent manner at higher stimulation rates. The voltage-dependence of inactivation was shifted in the hyperpolarizing direction, and recovery from inactivation was slowed by BBG. The most dramatic effect of BBG was to slow recovery from inactivation after long depolarizations, with 3 μM BBG increasing half-time for recovery (measured at -120 mV) from 24 to 854 ms after a 10-s step to 0 mV. These results were mimicked by a kinetic model in which BBG binds weakly to resting channels (K(d) = 170 μM) but tightly to fast-inactivated channels (K(d) = 5 μM) and even more tightly (K(d) = 0.2 μM) to slow-inactivated channels. In contrast to BBG, the structurally related food-coloring dye Brilliant Blue FCF had very little effect at concentrations up to 30 μM. These results show that BBG inhibits voltage-gated sodium channels at micromolar concentrations. Although BBG inhibition of sodium channels is less potent than inhibition of P2X7 receptors, there may be significant inhibition of sodium channels at BBG concentrations achieved in spinal cord or brain during experimental treatment of spinal cord injury or Huntington's disease. Considered as a sodium channel blocker, BBG is remarkably potent, acting with more than 10-fold greater potency than lacosamide, another blocker thought to bind to slow-inactivated channels.
Collapse
Affiliation(s)
- Sooyeon Jo
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | | |
Collapse
|
13
|
Thompson CH, Kahlig KM, George AL. SCN1A splice variants exhibit divergent sensitivity to commonly used antiepileptic drugs. Epilepsia 2011; 52:1000-9. [PMID: 21453355 DOI: 10.1111/j.1528-1167.2011.03040.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE A common genetic variant (rs3812718) in a splice donor consensus sequence within the neuronal sodium channel gene SCN1A (encoding Na(V) 1.1) modulates the proportion of transcripts incorporating either the canonical (5A) or alternative (5N) exon 5. A pharmacogenetic association has been reported whereby increased expression of exon 5N containing Na(V) 1.1 transcripts correlated with lower required doses of phenytoin in epileptics. We tested the hypothesis that SCN1A alternative splicing affects the pharmacology of Na(V) 1.1 channels. METHODS To directly examine biophysical and pharmacologic differences between the exon 5 splice variants, we performed whole-cell patch clamp recording of tsA201 cells transiently coexpressing either Na(V) 1.1-5A or Na(V) 1.1-5N with the β1 and β2 accessory subunits. We examined tonic inhibition and use-dependent inhibition of Na(V) 1.1 splice isoforms by phenytoin, carbamazepine, and lamotrigine. We also examined the effects of phenytoin and lamotrigine on channel biophysical properties and determined concentration-response relationships for both splice variants. KEY FINDINGS We observed no significant differences in voltage dependence of activation, steady-state inactivation, and recovery from inactivation between splice variants. However, Na(V) 1.1-5N channels exhibited enhanced tonic block by phenytoin and lamotrigine compared to Na(V) 1.1-5A. In addition, Na(V) 1.1-5N exhibited enhanced use-dependent block by phenytoin and lamotrigine across a range of stimulation frequencies and concentrations. Phenytoin and lamotrigine induced shifts in steady-state inactivation and recovery from fast inactivation for both splice isoforms. No splice isoform differences were observed for channel inhibition by carbamazepine. SIGNIFICANCE These results suggest Na(V) 1.1 channels containing exon 5N are more sensitive to the commonly used antiepileptic drugs phenytoin and lamotrigine.
Collapse
|
14
|
Iwamoto T, Takasugi Y, Higashino H, Ito H, Koga Y, Nakao S. Antinociceptive action of carbamazepine on thermal hypersensitive pain at spinal level in a rat model of adjuvant-induced chronic inflammation. J Anesth 2010; 25:78-86. [PMID: 21113631 DOI: 10.1007/s00540-010-1046-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 10/25/2010] [Indexed: 12/14/2022]
Abstract
PURPOSE Systemic carbamazepine, a voltage-gated sodium channel blocker, has been reported to dose-dependently reduce inflammatory hyperalgesia. However, the antinociceptive effects of carbamazepine on the spinal cord in inflammatory conditions are unclear. The aim of the present study was to evaluate the antinociceptive effects of carbamazepine on the spinal cord in a chronic inflammatory condition. METHODS In Sprague-Dawley rats, a chronic inflammatory condition was induced by complete Freund's adjuvant (CFA) inoculation into the tail. Tail flick (TF) latencies were measured following intraperitoneal carbamazepine, or intrathecal carbamazepine or tetrodotoxin injection in intact rats and in the chronic inflammatory rats. From the values of TF latency at 60 min after drug injection, the effective dose required to produce 50% response (ED(50)) of each drug was derived. RESULTS Carbamazepine attenuated thermal responses with both systemic and intrathecal administration. The effect was more evident in rats with chronic inflammation than in intact rats; the ED(50s) of intraperitoneal carbamazepine in intact and inflamed rats were 12.39 and 1.54 mg/kg, and those of intrathecal carbamazepine were 0.311 and 0.048 nmol, respectively. Intrathecal tetrodotoxin also clearly inhibited the response, with ED(50s) of 1.006 pmol in intact rats and 0.310 pmol in inflamed rats. The relative potencies of intrathecal carbamazepine versus tetrodotoxin for inhibition were approximately 1:150-1:300 in intact and inflamed rats. CONCLUSION These results indicate that the inhibition of voltage-gated sodium channels, at least tetrodotoxin-sensitive channels, may contribute to the antinociceptive effect of carbamazepine on CFA-induced inflammatory pain, since lower doses of intrathecal carbamazepine and tetrodotoxin attenuated thermal responses to a greater extent in inflamed rats than in intact rats.
Collapse
Affiliation(s)
- Tatsushige Iwamoto
- Department of Anesthesiology, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | | | | | | | | | | |
Collapse
|
15
|
Yau HJ, Baranauskas G, Martina M. Flufenamic acid decreases neuronal excitability through modulation of voltage-gated sodium channel gating. J Physiol 2010; 588:3869-82. [PMID: 20724367 DOI: 10.1113/jphysiol.2010.193037] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The electrophysiological phenotype of individual neurons critically depends on the biophysical properties of the voltage-gated channels they express. Differences in sodium channel gating are instrumental in determining the different firing phenotypes of pyramidal cells and interneurons; moreover, sodium channel modulation represents an important mechanism of action for many widely used CNS drugs. Flufenamic acid (FFA) is a non-steroidal anti-inflammatory drug that has been long used as a blocker of calcium-dependent cationic conductances. Here we show that FFA inhibits voltage-gated sodium currents in hippocampal pyramidal neurons; this effect is dose-dependent with IC(50) = 189 μm. We used whole-cell and nucleated patch recordings to investigate the mechanisms of FFA modulation of TTX-sensitive voltage-gated sodium current. Our data show that flufenamic acid slows down the inactivation process of the sodium current, while shifting the inactivation curve ~10 mV toward more hyperpolarized potentials. The recovery from inactivation is also affected in a voltage-dependent way, resulting in slower recovery at hyperpolarized potentials. Recordings from acute slices demonstrate that FFA reduces repetitive- and abolishes burst-firing in CA1 pyramidal neurons. A computational model based on our data was employed to better understand the mechanisms of FFA action. Simulation data support the idea that FFA acts via a novel mechanism by reducing the voltage dependence of the sodium channel fast inactivation rates. These effects of FFA suggest that it may be an effective anti-epileptic drug.
Collapse
Affiliation(s)
- Hau-Jie Yau
- Department of Physiology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | | | | |
Collapse
|
16
|
Yang YC, Hsieh JY, Kuo CC. The external pore loop interacts with S6 and S3-S4 linker in domain 4 to assume an essential role in gating control and anticonvulsant action in the Na(+) channel. ACTA ACUST UNITED AC 2009; 134:95-113. [PMID: 19635852 PMCID: PMC2717694 DOI: 10.1085/jgp.200810158] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Carbamazepine, phenytoin, and lamotrigine are widely prescribed anticonvulsants in neurological clinics. These drugs bind to the same receptor site, probably with the diphenyl motif in their structure, to inhibit the Na+ channel. However, the location of the drug receptor remains controversial. In this study, we demonstrate close proximity and potential interaction between an external aromatic residue (W1716 in the external pore loop) and an internal aromatic residue (F1764 in the pore-lining part of the sixth transmembrane segment, S6) of domain 4 (D4), both being closely related to anticonvulsant and/or local anesthetic binding to the Na+ channel. Double-mutant cycle analysis reveals significant cooperativity between the two phenyl residues for anticonvulsant binding. Concomitant F1764C mutation evidently decreases the susceptibility of W1716C to external Cd2+ and membrane-impermeable methanethiosulfonate reagents. Also, the W1716E/F1764R and G1715E/F1764R double mutations significantly alter the selectivity for Na+ over K+ and markedly shift the activation curve, respectively. W1716 and F1764 therefore very likely form a link connecting the outer and inner compartments of the Na+ channel pore (in addition to the selectivity filter). Anticonvulsants and local anesthetics may well traverse this “S6 recess” without trespassing on the selectivity filter. Furthermore, we found that Y1618K, a point mutation in the S3-4 linker (the extracellular extension of D4S4), significantly alters the consequences of carbamazepine binding to the Na+ channel. The effect of Y1618K mutation, however, is abolished by concomitant point mutations in the vicinity of Y1618, but not by those in the internally located inactivation machinery, supporting a direct local rather than a long-range allosteric action. Moreover, Y1618 could interact with D4 pore residues W1716 and L1719 to have a profound effect on both channel gating and anticonvulsant action. We conclude that there are direct interactions among the external S3-4 linker, the external pore loop, and the internal S6 segment in D4, making the external pore loop a pivotal point critically coordinating ion permeation, gating, and anticonvulsant binding in the Na+ channel.
Collapse
Affiliation(s)
- Ya-Chin Yang
- Department of Life Science, Chang-Gung University, Tao-Yuan, Taiwan
| | | | | |
Collapse
|
17
|
He YL, Zhan XQ, Yang G, Sun J, Mei YA. Amoxapine inhibits the delayed rectifier outward K+ current in mouse cortical neurons via cAMP/protein kinase A pathways. J Pharmacol Exp Ther 2009; 332:437-45. [PMID: 19915071 DOI: 10.1124/jpet.109.159160] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ion channels are known to be modulated by antidepressant drugs, but the molecular mechanisms are not known. We have shown that the antidepressant drug amoxapine suppresses rectifier outward K(+) (I(K)) currents in mouse cortical neurons. At a concentration of 10 to 500 muM, amoxapine reversibly inhibited I(K) in a dose-dependent manner and modulated both steady-state activation and inactivation properties. The application of forskolin or dibutyryl cAMP mimicked the inhibitory effect of amoxapine on I(K) and abolished further inhibition by amoxapine. N-[2-(p-Bromocinnamylamino)ethyl]-5-iso-quinolinesulphonamide (H-89), a protein kinase A (PKA) inhibitor, augmented I(K) amplitudes and completely eliminated amoxapine inhibition of I(K). Amoxapine was also found to significantly increase intracellular cAMP levels. The effects of amoxapine on I(K) were abolished by preincubation with 5-hydroxytryptamine (5-HT) and the antagonists of 5-HT(2) receptor. Moreover, intracellular application of guanosine 5'-[gammathio]-triphosphate increased I(K) amplitudes and prevented amoxapine-induced inhibition. The selective Kv2.1 subunit blocker Jingzhaotoxin-III reduced I(K) amplitudes by 30% and also significantly abolished the inhibitory effect of amoxapine. Together these results suggest that amoxapine inhibits I(K) in mouse cortical neurons by cAMP/PKA-dependent pathway associated with the 5-HT receptor, and suggest that the Kv2.1 alpha-subunit may be the target for this inhibition.
Collapse
Affiliation(s)
- Yan-Lin He
- nstitutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
18
|
Fischer TZ, Gilmore ES, Estacion M, Eastman E, Taylor S, Melanson M, Dib-Hajj SD, Waxman SG. A novel Nav1.7 mutation producing carbamazepine-responsive erythromelalgia. Ann Neurol 2009; 65:733-41. [PMID: 19557861 DOI: 10.1002/ana.21678] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Human and animal studies have shown that Na(v)1.7 sodium channels, which are preferentially expressed within nociceptors and sympathetic neurons, play a major role in inflammatory and neuropathic pain. Inherited erythromelalgia (IEM) has been linked to gain-of-function mutations of Na(v)1.7. We now report a novel mutation (V400M) in a three-generation Canadian family in which pain is relieved by carbamazepine (CBZ). METHODS We extracted genomic DNA from blood samples of eight members of the family, and the sequence of SCN9A coding exons was compared with the reference Na(v)1.7 complementary DNA. Wild-type Na(v)1.7 and V400M cell lines were then analyzed using whole-cell patch-clamp recording for changes in activation, deactivation, steady-state inactivation, and ramp currents. RESULTS Whole-cell patch-clamp studies of V400M demonstrate changes in activation, deactivation, steady-state inactivation, and ramp currents that can produce dorsal root ganglia neuron hyperexcitability that underlies pain in these patients. We show that CBZ, at concentrations in the human therapeutic range, normalizes the voltage dependence of activation and inactivation of this inherited erythromelalgia mutation in Na(v)1.7 but does not affect these parameters in wild-type Na(v)1.7. INTERPRETATION Our results demonstrate a normalizing effect of CBZ on mutant Na(v)1.7 channels in this kindred with CBZ-responsive inherited erythromelalgia. The selective effect of CBZ on the mutant Na(v)1.7 channel appears to explain the ameliorative response to treatment in this kindred. Our results suggest that functional expression and pharmacological studies may provide mechanistic insights into hereditary painful disorders.
Collapse
Affiliation(s)
- Tanya Z Fischer
- Department of Neurology, Yale University School of Medicine, New Haven, CT 16510, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Analysis of the Kinetics of Blockade of Tetrodotoxin-Sensitive and Tetrodotoxin-Resistant Sodium Channels Induced by an Analgesic, D57, in Neurons of the Rat Afferent Ganglia. NEUROPHYSIOLOGY+ 2009. [DOI: 10.1007/s11062-009-9056-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Bupivacaine blocks N-type inactivating Kv channels in the open state: no allosteric effect on inactivation kinetics. Biophys J 2008; 95:5138-52. [PMID: 18790854 DOI: 10.1529/biophysj.108.130518] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Local anesthetics bind to ion channels in a state-dependent manner. For noninactivating voltage-gated K channels the binding mainly occurs in the open state, while for voltage-gated inactivating Na channels it is assumed to occur mainly in inactivated states, leading to an allosterically caused increase in the inactivation probability, reflected in a negative shift of the steady-state inactivation curve, prolonged recovery from inactivation, and a frequency-dependent block. How local anesthetics bind to N-type inactivating K channels is less explored. In this study, we have compared bupivacaine effects on inactivating (Shaker and K(v)3.4) and noninactivating (Shaker-IR and K(v)3.2) channels, expressed in Xenopus oocytes. Bupivacaine was found to block these channels time-dependently without shifting the steady-state inactivation curve markedly, without a prolonged recovery from inactivation, and without a frequency-dependent block. An analysis, including computational testing of kinetic models, suggests binding to the channel mainly in the open state, with affinities close to those estimated for corresponding noninactivating channels (300 and 280 microM for Shaker and Shaker-IR, and 60 and 90 microM for K(v)3.4 and K(v)3.2). The similar magnitudes of K(d), as well as of blocking and unblocking rate constants for inactivating and noninactivating Shaker channels, most likely exclude allosteric interactions between the inactivation mechanism and the binding site. The relevance of these results for understanding the action of local anesthetics on Na channels is discussed.
Collapse
|
22
|
Williams WR. Voltage-gated Na+ channel ligands and ATP: relative molecular similarity and implications for channel function. J Pharm Pharmacol 2006; 58:1235-41. [PMID: 16945182 DOI: 10.1211/jpp.58.9.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The voltage-gated sodium channel (VGNC) is targeted by naturally occurring ligands and drugs of diverse structure. ATP modulates VGNC current in-vitro but is given little prominence in models describing channel function. This computational study uses superimposition and molecular fitting to investigate relative molecular similarity within the structures of ATP and VGNC ligands. A motif of 3 linked atoms (C-N-C) in the adenine ring of ATP satisfies the fitting of a wide range of anticonvulsant structures. An alternative group (N-C-N) provides one fitting motif for the ester and amide groups of local anaesthetic drugs; protonated amine and aromatic groups in the same conformers fit to a second motif in the adenine ring. Analogous structures from other drug classes with VGNC blocking activity give the same molecular fits to ATP. Structures fitted to the adenine ring of ATP occlude the intra-molecular space between the nucleoside and triphosphate chain in approximation to their established blocking, activating or neutral effects on Na+ current. The findings are discussed in terms of drug preferences for VGNC states and channel requirements for ATP.
Collapse
Affiliation(s)
- W R Williams
- School of Care Sciences, University of Glamorgan, Pontypridd CF37 1DL, UK.
| |
Collapse
|
23
|
Lenkowski PW, Batts TW, Smith M, Ko SH, Jones PJ, Taylor CH, McCusker AK, Davis GC, Hartmann HA, White HS, Brown ML, Patel MK. A pharmacophore derived phenytoin analogue with increased affinity for slow inactivated sodium channels exhibits a desired anticonvulsant profile. Neuropharmacology 2006; 52:1044-54. [PMID: 17174360 PMCID: PMC2517177 DOI: 10.1016/j.neuropharm.2006.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 10/20/2006] [Accepted: 11/01/2006] [Indexed: 01/15/2023]
Abstract
Phenytoin (DPH) is a clinically useful sodium (Na) channel blocker with efficacy against partial and generalized seizures. We have developed a novel hydantoin compound (HA) using comparative molecular field analysis (CoMFA) and evaluated its effects on hNa(v)1.2 channels. Both DPH and HA demonstrated affinity for resting (K(r)=13.9microM for HA, K(r)=464microM for DPH) and slow inactivated channels (K(I)=975nM for HA, K(I)=20.6microM for DPH). However, HA also exhibited an affinity for fast inactivated channels (K(I)=2.5microM) and shifted the V(1/2) for activation in the depolarizing direction. Furthermore, HA exhibited profound use dependent block at both 5 and 10Hz stimulation frequencies. In the 6Hz seizure model (32mA) HA had an ED(50) of 47.1mg/kg and a TD(50) of 131mg/kg (protective index (PI)=2.8). In comparison, the ED(50) for DPH was approximately 27.5mg/kg with a TD(50) of 35.6mg/kg (PI approximately 1.3). These findings provide evidence for the utility of CoMFA in the design of novel anticonvulsants and support the hypothesis that states selectivity plays an important role in achieving optimal protection with minimal side effects.
Collapse
Affiliation(s)
- Paul W. Lenkowski
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Timothy W. Batts
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Misty Smith
- Anticonvulsant Drug Development (ADD) Program (M.S-Y, H.S.W), Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, 84108
| | - Seong-Hoon Ko
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Paulianda J. Jones
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Catherine H. Taylor
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Ashley K. McCusker
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Gary C. Davis
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Hali A. Hartmann
- Medical Biotechnology Center (H.A.H), University of Maryland Biotechnology Institute, Baltimore, Maryland, 21201
| | - H. Steve White
- Anticonvulsant Drug Development (ADD) Program (M.S-Y, H.S.W), Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, 84108
| | - Milton L. Brown
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Manoj K. Patel
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia, 22908, USA
- Corresponding Author: Manoj K. Patel, Dept. Anesthesiology, Box 800710, University of Virginia Health System, Charlottesville, Virginia, 22908-0710, USA. Tel: +1 434 924 9693; Fax: +1 434 924 2105 ()
| |
Collapse
|
24
|
Eto K, Arimura Y, Mizuguchi H, Nishikawa M, Noda M, Ishibashi H. Modulation of ATP-induced inward currents by docosahexaenoic acid and other fatty acids in rat nodose ganglion neurons. J Pharmacol Sci 2006; 102:343-6. [PMID: 17099292 DOI: 10.1254/jphs.sc0060053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The effects of docosahexaenoic acid (DHA) and other fatty acids on P2X-receptor-mediated inward currents in rat nodose ganglion neurons were studied using the nystatin perforated patch-clamp technique. DHA accelerated the desensitization rate of the ATP-induced current. DHA showed use-dependent inhibition of the peak ATP-induced current. Other polyunsaturated fatty acids, such as arachidonic acid and eicosapentaenoic acid, displayed a similar use-dependent inhibition. The inhibitory effects of saturated fatty acids including palmitic acid and arachidic acid were weaker than those of polyunsaturated fatty acids. The results suggest that fatty acids may modulate the P2X receptor-mediated response when the channel is in the open-state.
Collapse
Affiliation(s)
- Kei Eto
- Department of Physiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Lenkey N, Karoly R, Kiss JP, Szasz BK, Vizi ES, Mike A. The Mechanism of Activity-Dependent Sodium Channel Inhibition by the Antidepressants Fluoxetine and Desipramine. Mol Pharmacol 2006; 70:2052-63. [PMID: 16985186 DOI: 10.1124/mol.106.026419] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The effect of monoamine uptake inhibitor-type antidepressants on sodium channels of hippocampal neurons was investigated. Members of the tricyclic group of antidepressants are known to modify multiple targets, including sodium channels, whereas selective serotonin-reuptake inhibitors (SSRIs) are regarded as highly selective compounds, and their effect on sodium channels was not investigated in detail. In this study, a representative member of each group was chosen: the tricyclic antidepressant desipramine and the SSRI fluoxetine. The drugs were roughly equipotent use-dependent inhibitors of sodium channels, with IC(50) values approximately 100 microM at -150 mV holding potential, and approximately 1 microM at -60 mV. We suggest that therapeutic concentrations of antidepressants affect neuronal information processing partly by direct, activity-dependent inhibition of sodium channels. As for the mechanism of inhibition, use-dependent inhibition by antidepressants was believed to be due to a preferential affinity to the fast-inactivated state. Using a voltage and perfusion protocol by which relative affinities to fast-versus slow-inactivated states could be assessed, we challenged this view and found that the affinity of both drugs to slowinactivated state(s) was higher. We propose a different mechanism of action for these antidepressants, in which slow rather than fast inactivation plays the dominant role. This mechanism is similar but not equivalent with the novel mechanism of usedependent sodium channel inhibition previously described by our group (Neuroscience 125:1019-1028, 2004; Neuroreport 14:1945-1949, 2003). Our results suggest that different drugs can produce use-dependent sodium channel inhibition by different mechanisms.
Collapse
Affiliation(s)
- N Lenkey
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
26
|
Cardenas CA, Cardenas CG, de Armendi AJ, Scroggs RS. Carbamazepine interacts with a slow inactivation state of NaV1.8-like sodium channels. Neurosci Lett 2006; 408:129-34. [PMID: 16978779 DOI: 10.1016/j.neulet.2006.08.070] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Revised: 08/10/2006] [Accepted: 08/29/2006] [Indexed: 10/24/2022]
Abstract
Carbamazepine was tested on high-threshold TTX-resistant Na+ currents (TTX-R-currents), evoked from acutely isolated rat dorsal root ganglion (DRG) cells. Under control conditions, the TTX-R-currents recorded from different DRG cells varied greatly regarding use-dependent inactivation (TTX-R-current UDI), measured as the percent decrease in current amplitude induced by changing the current activation rate from 0.1 Hz to 1.0 Hz. Also, when TTX-R-currents were evoked at 0.1 Hz from a holding potential (hp) of -60 mV, a larger fraction of TTX-R-channels resided tonically in a slow inactivation state in DRG cells with more TTX-R-current UDI versus those with less TTX-R-current UDI. The block of TTX-R-currents evoked from hp -60 mV by 100-microM carbamazepine and the EC50 for carbamazepine block was positively correlated with TTX-R-current UDI. The slope factors estimated for the concentration-response curves averaged 0.68, suggesting the presence of low and high affinity sites. Fitting the data with a two-site binding isotherm gave estimates of 30 microM and 760 microM for the EC50s of the high and low affinity sites, respectively. The fraction of the total fit attributed to the high affinity site was positively correlated with TTX-R-current UDI. Carbamazepine increased the fast and slow time constants for recovery from inactivation and the fraction of the fit attributed to the slow time constant. These data suggest that carbamazepine interacts with a slow inactivation state of TTX-R-channels. This particular mechanism might be exploited in future research aimed at developing pain medications that selectively block Na(V)1.8 channels or Na+ channels in general.
Collapse
Affiliation(s)
- Carlos A Cardenas
- Division of Anesthesiology, St. Jude Children's Research Hospital, Memphis, TN 38163, USA
| | | | | | | |
Collapse
|
27
|
Benjamin ER, Pruthi F, Olanrewaju S, Shan S, Hanway D, Liu X, Cerne R, Lavery D, Valenzano KJ, Woodward RM, Ilyin VI. Pharmacological characterization of recombinant N-type calcium channel (Cav2.2) mediated calcium mobilization using FLIPR. Biochem Pharmacol 2006; 72:770-82. [PMID: 16844100 DOI: 10.1016/j.bcp.2006.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2006] [Revised: 06/04/2006] [Accepted: 06/06/2006] [Indexed: 11/24/2022]
Abstract
The N-type voltage-gated calcium channel (Ca(v)2.2) functions in neurons to regulate neurotransmitter release. It comprises a clinically relevant target for chronic pain. We have validated a calcium mobilization approach to assessing Ca(v)2.2 pharmacology in two stable Ca(v)2.2 cell lines: alpha1(B), alpha2delta, beta(3)-HEK-293 and alpha1(B), beta(3)-HEK-293. Ca(v)2.2 channels were opened by addition of KCl and Ca(2+) mobilization was measured by Fluo-4 fluorescence on a fluorescence imaging plate reader (FLIPR(96)). Ca(v)2.2 expression and biophysics were confirmed by patch-clamp electrophysiology (EP). Both cell lines responded to KCl with adequate signal-to-background. Signals from both cell lines were inhibited by omega-conotoxin (ctx)-MVIIa and omega-conotoxin (ctx)-GVIa with IC(50) values of 1.8 and 1nM, respectively, for the three-subunit stable, and 0.9 and 0.6nM, respectively, for the two-subunit stable. Other known Ca(v)2.2 blockers were characterized including cadmium, flunarizine, fluspirilene, and mibefradil. IC(50) values correlated with literature EP-derived values. Novel Ca(v)2.2 pharmacology was identified in classes of compounds with other primary pharmacological activities, including Na(+) channel inhibitors and antidepressants. Novel Na(+) channel compounds with high potency at Ca(v)2.2 were identified in the phenoxyphenyl pyridine, phenoxyphenyl pyrazole, and other classes. The highest potency at Ca(v)2.2 tricyclic antidepressant identified was desipramine.
Collapse
Affiliation(s)
- Elfrida R Benjamin
- Purdue Pharma Discovery Research, 6 Cedarbrook Drive, Cranbury, NJ 08512, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ko SH, Jochnowitz N, Lenkowski PW, Batts TW, Davis GC, Martin WJ, Brown ML, Patel MK. Reversal of neuropathic pain by α-hydroxyphenylamide: A novel sodium channel antagonist. Neuropharmacology 2006; 50:865-73. [PMID: 16464480 DOI: 10.1016/j.neuropharm.2005.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 12/06/2005] [Accepted: 12/14/2005] [Indexed: 01/31/2023]
Abstract
Sodium (Na) channel blockers are known to possess antihyperalgesic properties. We have designed and synthesized a novel Na channel antagonist, alpha-hydroxyphenylamide, and determined its ability to inhibit both TTX-sensitive (TTX-s) and TTX-resistant (TTX-r) Na currents from small dorsal root ganglion (DRG) neurons. alpha-Hydroxyphenylamide tonically inhibited both TTX-s and TTX-r Na currents yielding an IC(50) of 8.2+/-2.2 microM (n=7) and 28.9+/-1.8 microM (n=8), respectively. In comparison, phenytoin was less potent inhibiting TTX-s and TTX-r currents by 26.2+/-4.0% (n=8) and 25.5+/-2.0%, respectively, at 100 microM. alpha-Hydroxyphenylamide (10 microM) also shifted equilibrium gating parameters of TTX-s Na channels to greater hyperpolarized potentials, slowed recovery from inactivation, accelerated the development of inactivation and exhibited use-dependent block. In the chronic constriction injury (CCI) rat model of neuropathic pain, intraperitoneal administration of alpha-hydroxyphenylamide attenuated the hyperalgesia by 53% at 100mg/kg, without affecting motor coordination in the Rotorod test. By contrast, the reduction in pain behavior produced by phenytoin (73%; 100mg/kg) was associated with significant motor impairment. In summary, we report that alpha-hydroxyphenylamide, a sodium channel antagonist, exhibits antihyperalgesic properties in a rat model of neuropathic pain, with favorable sedative and ataxic side effects compared with phenytoin.
Collapse
Affiliation(s)
- Seong-Hoon Ko
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, Box 800710, Charlottesville, VA 22908-0710, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Alvarez-Silva S, Alvarez-Silva I, Alvarez-Rodriguez J, Perez-Echeverria MJ, Campayo-Martinez A, Rodriguez-Fernandez FL. Epileptic consciousness: concept and meaning of aura. Epilepsy Behav 2006; 8:527-33. [PMID: 16510316 DOI: 10.1016/j.yebeh.2005.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Revised: 12/15/2005] [Accepted: 12/21/2005] [Indexed: 10/25/2022]
Abstract
This research is based on previous publications that have analyzed certain neuropsychological phenomena that always have the same characteristic clinical features: a vivid experience of sudden onset and automatic development, accompanied by an intense sensation of strangeness. When these automatisms are accompanied by only mental symptoms, the designation paroxysmal psychic automatisms (PPAs) is proposed, and they should be interpreted as partial seizures (PSs) with a psychic content whenever they clearly exhibit the four features of suddenness, passivity, intensity, and strangeness. This interpretation is based on the existence of a wealth of scientific literature indicating an overlap between PPAs and PSs; moreover, bibliographic reviews indicate that the clinical signs just defined as characterizing PPAs are precisely those defining the epileptic consciousness.
Collapse
|
30
|
Huang CJ, Harootunian A, Maher MP, Quan C, Raj CD, McCormack K, Numann R, Negulescu PA, González JE. Characterization of voltage-gated sodium-channel blockers by electrical stimulation and fluorescence detection of membrane potential. Nat Biotechnol 2006; 24:439-46. [PMID: 16550174 DOI: 10.1038/nbt1194] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Accepted: 01/05/2006] [Indexed: 11/09/2022]
Abstract
Voltage-gated ion channels regulate many physiological functions and are targets for a number of drugs. Patch-clamp electrophysiology is the standard method for measuring channel activity because it fulfils the requirements for voltage control, repetitive stimulation and high temporal resolution, but it is laborious and costly. Here we report an electro-optical technology and automated instrument, called the electrical stimulation voltage ion probe reader (E-VIPR), that measures the activity of voltage-gated ion channels using extracellular electrical field stimulation and voltage-sensitive fluorescent probes. We demonstrate that E-VIPR can sensitively detect drug potency and mechanism of block on the neuronal human type III voltage-gated sodium channel expressed in human embryonic kidney cells. Results are compared with voltage-clamp and show that E-VIPR provides sensitive and information-rich compound blocking activity. Furthermore, we screened approximately 400 drugs and observed sodium channel-blocking activity for approximately 25% of them, including the antidepressants sertraline (Zoloft) and paroxetine (Paxil).
Collapse
Affiliation(s)
- Chien-Jung Huang
- Vertex Pharmaceuticals Incorporated, 11010 Torreyana Road, San Diego, California 92121, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Webster RG, Brain KL, Wilson RH, Grem JL, Vincent A. Oxaliplatin induces hyperexcitability at motor and autonomic neuromuscular junctions through effects on voltage-gated sodium channels. Br J Pharmacol 2006; 146:1027-39. [PMID: 16231011 PMCID: PMC1751225 DOI: 10.1038/sj.bjp.0706407] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Oxaliplatin, an effective cytotoxic treatment in combination with 5-fluorouracil for colorectal cancer, is associated with sensory, motor and autonomic neurotoxicity. Motor symptoms include hyperexcitability while autonomic effects include urinary retention, but the cause of these side-effects is unknown. We examined the effects on motor nerve function in the mouse hemidiaphragm and on the autonomic system in the vas deferens. In the mouse diaphragm, oxaliplatin (0.5 mM) induced multiple endplate potentials (EPPs) following a single stimulus, and was associated with an increase in spontaneous miniature EPP frequency. In the vas deferens, spontaneous excitatory junction potential frequency was increased after 30 min exposure to oxaliplatin; no changes in resting Ca(2+) concentration in nerve terminal varicosities were observed, and recovery after stimuli trains was unaffected. In both tissues, an oxaliplatin-induced increase in spontaneous activity was prevented by the voltage-gated Na(+) channel blocker tetrodotoxin (TTX). Carbamazepine (0.3 mM) also prevented multiple EPPs and the increase in spontaneous activity in both tissues. In diaphragm, beta-pompilidotoxin (100 microM), which slows Na(+) channel inactivation, induced multiple EPPs similar to oxaliplatin's effect. By contrast, blockers of K(+) channels (4-aminopyridine and apamin) did not replicate oxaliplatin-induced hyperexcitability in the diaphragm. The prevention of hyperexcitability by TTX blockade implies that oxaliplatin acts on nerve conduction rather than by effecting repolarisation. The similarity between beta-pompilidotoxin and oxaliplatin suggests that alteration of voltage-gated Na(+) channel kinetics is likely to underlie the acute neurotoxic actions of oxaliplatin.
Collapse
Affiliation(s)
- Richard G Webster
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS
- Author for correspondence:
| | - Keith L Brain
- University Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Richard H Wilson
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Jean L Grem
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Angela Vincent
- Neurosciences Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS
| |
Collapse
|
32
|
Fuller MD, Zhang ZR, Cui G, McCarty NA. The block of CFTR by scorpion venom is state-dependent. Biophys J 2005; 89:3960-75. [PMID: 16183882 PMCID: PMC1366962 DOI: 10.1529/biophysj.105.060731] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Accepted: 09/13/2005] [Indexed: 02/01/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) adenosine triphosphate-dependent chloride channels are expressed in epithelial cells and are associated with a number of genetic disorders, including cystic fibrosis. Venom of the scorpion Leirus quinquestriatus hebraeus reversibly inhibits CFTR when applied to its cytoplasmic surface. To examine the state-dependence of inhibition we recorded wild-type and mutant CFTR channel currents using inside-out membrane patches from Xenopus oocytes. Application of either venom or diphenylamine-2-carboxylate to channels that were either activated (open) or resting (closed) indicate primarily closed state-dependent inhibition of CFTR by venom, whereas diphenylamine-2-carboxylate showed no state-dependence of block. Efficacy of venom-mediated macroscopic current inhibition was inversely related to channel activity. Analysis of single-channel and macropatch data indicated that venom could either inhibit channel opening, if it binds during an interburst closed state or in the absence of cytosolic adenosine triphosphate, or introduce new intraburst closed states, if it binds during an open event. The on-rate of venom binding for intraburst block could be modulated by changing CFTR activity with vanadate or adenylyl-imidodiphosphate, or by introducing the Walker A mutation K1250A. These findings represent the first description of state-dependent inhibition of CFTR and suggest that the active toxin could be used as a tool to study the conformational changes that occur during CFTR gating.
Collapse
Affiliation(s)
- Matthew D Fuller
- Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30332-0230, USA
| | | | | | | |
Collapse
|
33
|
Benjamin ER, Pruthi F, Olanrewaju S, Ilyin VI, Crumley G, Kutlina E, Valenzano KJ, Woodward RM. State-Dependent Compound Inhibition of Nav1.2 Sodium Channels Using the FLIPR VmDye: On-Target and Off-Target Effects of Diverse Pharmacological Agents. ACTA ACUST UNITED AC 2005; 11:29-39. [PMID: 16234337 DOI: 10.1177/1087057105280918] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Voltage-gated sodiumchannels (NaChs) are relevant targets for pain, epilepsy, and a variety of neurological and cardiac disorders. Traditionally, it has been difficult to develop structure-activity relationships for NaCh inhibitors due to rapid channel kinetics and state-dependent compound interactions. Membrane potential (Vm)dyes in conjunctionwith a high-throughput fluorescence imaging plate reader (FLIPR) offer a satisfactory 1st-tier solution. Thus, the authors have developed a FLIPR Vmassay of rat Nav1.2NaCh. Channels were opened by addition of veratridine, and Vm dye responses were measured. The IC50 values from various structural classes of compounds were compared to the resting state binding constant (Kr)and inactivated state binding constant (Ki)obtained using patch-clamp electrophysiology (EP). The FLIPR values correlated with Ki but not Kr.FLIPRIC50 values fellwithin 0.1-to 1.5-fold of EPKi values, indicating that the assay generally reports use-dependent inhibition rather than resting state block. The Library of Pharmacologically Active Compounds (LOPAC, Sigma) was screened. Confirmed hits arose from diverse classes such as dopamine receptor antagonists, serotonin transport inhibitors, and kinase inhibitors. These data suggest that NaCh inhibition is inherent in a diverse set of biologically active molecules and may warrant counterscreening NaChs to avoid unwanted secondary pharmacology.
Collapse
|
34
|
Braun M, Chaloupka V. Carbamazepine induced pitch shift and octave space representation. Hear Res 2005; 210:85-92. [PMID: 16181754 DOI: 10.1016/j.heares.2005.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2005] [Accepted: 05/12/2005] [Indexed: 10/25/2022]
Abstract
Octave-circular pitch perception, the repetition of pitch scale qualities when surpassing the octave interval, has been observed in behavioral data from humans and monkeys, but the underlying anatomy and physiology is still unknown. Here we analyze octave circularity in a concert pianist with absolute pitch, both under medication with the neurotropic drug carbamazepine (CBZ) and without medication. Analysis of 4619 responses in a pitch identification task revealed an internal tone-scale representation, based on the norm-tone scale re A4=440 Hz, with an octave-circular pattern of strongly and weakly represented tones. CBZ caused a global down-shift of pitch (ca. 1 semitone at 500 Hz), but no down-shift of the octave-circular pattern of tone characteristics. This pattern was similar in the six tested octave ranges (32.7-2093 Hz), both under the control and the CBZ condition. Pattern repetition always occurred at octave intervals and did not reflect the stretched octaves of piano tuning. The results indicate that CBZ influences pitch detection peripheral of an octave-circular pitch representation. Thus they support previous evidence for pitch detection in the auditory midbrain and for octave-circular pitch mapping in the auditory thalamus.
Collapse
Affiliation(s)
- Martin Braun
- Neuroscience of Music, Gansbyn 14, S-67195 Klässbol, Sweden.
| | | |
Collapse
|
35
|
Ilyin VI, Hodges DD, Whittemore ER, Carter RB, Cai SX, Woodward RM. V102862 (Co 102862): a potent, broad-spectrum state-dependent blocker of mammalian voltage-gated sodium channels. Br J Pharmacol 2005; 144:801-12. [PMID: 15778702 PMCID: PMC1576061 DOI: 10.1038/sj.bjp.0706058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
1. 4-(4-Fluorophenoxy)benzaldehyde semicarbazone (V102862) was initially described as an orally active anticonvulsant with robust activity in a variety of rodent models of epilepsy. The mechanism of action was not known. We used whole-cell patch-clamp techniques to study the effects of V102862 on native and recombinant mammalian voltage-gated Na+ channels. 2. V102862 blocked Na+ currents (I(Na)) in acutely dissociated cultured rat hippocampal neurons. Potency increased with membrane depolarization, suggesting a state-dependent mechanism of inhibition. There was no significant effect on the voltage dependence of activation of I(Na). 3. The dissociation constant for the inactivated state (K(I)) was approximately 0.6 microM, whereas the dissociation constant for the resting state (K(R)) was >15 microM. 4. The binding to inactivated channels was slow, requiring a few seconds to reach steady state at -80 mV. 5. The mechanism of inhibition was characterized in more detail using human embryonic kidney-293 cells stably expressing rat brain type IIA Na+ (rNa(v)1.2) channels, a major Na+ channel alpha subunit in rat hippocampal neurons. Similar to hippocampal neurons, V102862 was a potent state-dependent blocker of rNa(v)1.2 channels with a K(I) of approximately 0.4 microM and K(R) approximately 30 microM. V102862 binding to inactivated channels was relatively slow (k(+) approximately = 1.7 microM(-1) s(-1)). V102862 shifted the steady-state availability curve in the hyperpolarizing direction and significantly retarded recovery of Na+ channels from inactivation. 6. These results suggest that inhibition of voltage-gated Na+ channels is a major mechanism underlying the anticonvulsant properties of V102862. Moreover, understanding the biophysics of the interaction may prove to be useful in designing a new generation of potent Na+ channel blocker therapeutics.
Collapse
Affiliation(s)
- Victor I Ilyin
- Discovery Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, NJ 08512-3612, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Yang YC, Kuo CC. An inactivation stabilizer of the Na+ channel acts as an opportunistic pore blocker modulated by external Na+. ACTA ACUST UNITED AC 2005; 125:465-81. [PMID: 15824190 PMCID: PMC2217501 DOI: 10.1085/jgp.200409156] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The Na+ channel is the primary target of anticonvulsants carbamazepine, phenytoin, and lamotrigine. These drugs modify Na+ channel gating as they have much higher binding affinity to the inactivated state than to the resting state of the channel. It has been proposed that these drugs bind to the Na+ channel pore with a common diphenyl structural motif. Diclofenac is a widely prescribed anti-inflammatory agent that has a similar diphenyl motif in its structure. In this study, we found that diclofenac modifies Na+ channel gating in a way similar to the foregoing anticonvulsants. The dissociation constants of diclofenac binding to the resting, activated, and inactivated Na+ channels are ∼880 μM, ∼88 μM, and ∼7 μM, respectively. The changing affinity well depicts the gradual shaping of a use-dependent receptor along the gating process. Most interestingly, diclofenac does not show the pore-blocking effect of carbamazepine on the Na+ channel when the external solution contains 150 mM Na+, but is turned into an effective Na+ channel pore blocker if the extracellular solution contains no Na+. In contrast, internal Na+ has only negligible effect on the functional consequences of diclofenac binding. Diclofenac thus acts as an “opportunistic” pore blocker modulated by external but not internal Na+, indicating that the diclofenac binding site is located at the junction of a widened part and an acutely narrowed part of the ion conduction pathway, and faces the extracellular rather than the intracellular solution. The diclofenac binding site thus is most likely located at the external pore mouth, and undergoes delicate conformational changes modulated by external Na+ along the gating process of the Na+ channel.
Collapse
Affiliation(s)
- Ya-Chin Yang
- Department of Physiology, National Taiwan University College of Medicine, Taipei
| | | |
Collapse
|
37
|
Ebel H, Hollstein M, Günther T. Differential effect of imipramine and related compounds on Mg2+ efflux from rat erythrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2005; 1667:132-40. [PMID: 15581848 DOI: 10.1016/j.bbamem.2004.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2004] [Revised: 09/13/2004] [Accepted: 09/15/2004] [Indexed: 11/25/2022]
Abstract
The effect of imipramine on Mg2+ efflux in NaCl medium (Na+/Mg2+ antiport), on Mg2+ efflux in choline.Cl medium (choline/Mg2+ antiport) and on Mg2+ efflux in sucrose medium (Cl- -coupled Mg2+ efflux) was investigated in rat erythrocytes. In non-Mg2+-loaded rat erythrocytes, imipramine stimulated Na+/Mg2+ antiport but inhibited choline/Mg2+ antiport and Cl- -coupled Mg2+ efflux. The same effect could be obtained by several other compounds structurally related to imipramine. These drugs contain a cyclic hydrophobic ring structure to which a four-membered secondary or tertiary amine side chain is attached. At a physiological pH, the amine side chain expresses a cationic choline-like structure. The inhibitory effect on choline/Mg2+ antiport is lost when the amine side chain is modified or abandoned, pointing to competition of the choline-like side chain with choline or another cation at the unspecific choline antiporter or at the Cl- -coupled Mg2+ efflux. Other related drugs either stimulated Na+/Mg2+ antiport and choline/Mg2+ antiport, or they were ineffective. For stimulation of Na+/Mg2+ antiport and choline/Mg2+ antiport, there is no specific common structural motif of the drugs tested. The effects of imipramine on Na+/Mg2+ antiport and choline/Mg2+ antiport are not mediated by PKCalpha but are caused by a direct reaction of imipramine with these transporters. By increasing the intracellular Mg2+ concentration, the stimulation of Na+/Mg2+ antiport at a physiological intracellular Mg2+ concentration changed to an inhibition of Na+/Mg2+ antiport. This effect can be explained by the hypothesis that Mg2+ loading induced an allosteric transition of the Mg2+/Mg2+ exchanger with low Na+/Mg2+ antiport capacity to the Na+/Mg2+ antiporter with high Na+/Mg2+ antiport capacity. Both forms of the Mg2+ exchanger may be differently affected by imipramine.
Collapse
Affiliation(s)
- H Ebel
- Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Institut für Klinische Physiologie, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | | | |
Collapse
|
38
|
Alvarez-Rodriguez J, Alvarez-Silva S, Alvarez-Silva I. Epilepsy and psychiatry: Automatic psychic paroxysms. Med Hypotheses 2005; 65:671-5. [PMID: 16002226 DOI: 10.1016/j.mehy.2005.03.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Accepted: 03/16/2005] [Indexed: 11/28/2022]
Abstract
This study analyzes diverse psychic phenomena which, although always occurring with very characteristic clinical features, are sometimes diagnosed as epilepsy and sometimes as symptoms of different psychiatric disorders depending on the availability of an electroencephalogram. It is posited that these phenomena, whenever they are accompanied by the features characteristic of an epileptic consciousness (suddenness, automatic nature, great intensity and a strong sensation of strangeness) should be diagnosed as partial seizures with a psychic content, regardless of the availability of an EEG. The co-occurrence of these four clinical signs, which are relatively simple to objectivize, is a more reliable clinical criterion for diagnosis than a transcraneal electroencephalogram, which, as is known, is of little value in measuring the electrical activity of partial seizures. Moreover, an interpretation of this type makes it possible to reconcile scientific data from various neurosciences which thus far have seemed contradictory.
Collapse
Affiliation(s)
- J Alvarez-Rodriguez
- Servicio de Psiquiatria, Hospital de Leon, Altos de Nava s/n, Leon 24890, Spain.
| | | | | |
Collapse
|
39
|
García-Ferreiro RE, Kerschensteiner D, Major F, Monje F, Stühmer W, Pardo LA. Mechanism of block of hEag1 K+ channels by imipramine and astemizole. ACTA ACUST UNITED AC 2004; 124:301-17. [PMID: 15365094 PMCID: PMC2233905 DOI: 10.1085/jgp.200409041] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ether à go-go (Eag; KV10.1) voltage-gated K+ channels have been detected in cancer cell lines of diverse origin and shown to influence their rate of proliferation. The tricyclic antidepressant imipramine and the antihistamine astemizole inhibit the current through Eag1 channels and reduce the proliferation of cancer cells. Here we describe the mechanism by which both drugs block human Eag1 (hEag1) channels. Even if both drugs differ in their affinity for hEag1 channels (IC50s are approximately 2 microM for imipramine and approximately 200 nM for astemizole) and in their blocking kinetics, both drugs permeate the membrane and inhibit the hEag1 current by selectively binding to open channels. Furthermore, both drugs are weak bases and the IC50s depend on both internal an external pH, suggesting that both substances cross the membrane in their uncharged form and act from inside the cell in their charged forms. Accordingly, the block by imipramine is voltage dependent and antagonized by intracellular TEA, consistent with imipramine binding in its charged form to a site located close to the inner end of the selectivity filter. Using inside- and outside-out patch recordings, we found that a permanently charged, quaternary derivative of imipramine (N-methyl-imipramine) only blocks channels from the intracellular side of the membrane. In contrast, the block by astemizole is voltage independent. However, as astemizole competes with imipramine and intracellular TEA for binding to the channel, it is proposed to interact with an overlapping intracellular binding site. The significance of these findings, in the context of structure-function of channels of the eag family is discussed.
Collapse
Affiliation(s)
- Rafael E García-Ferreiro
- Abteilung Molekulare Biologie Neuronaler Signale, Max-Planck Institut für Experimentelle Medizin, 37075 Göttingen, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Pásek M, Simurda J. Quantitative modelling of interaction of propafenone with sodium channels in cardiac cells. Med Biol Eng Comput 2004; 42:151-7. [PMID: 15125143 DOI: 10.1007/bf02344625] [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: 01/11/2023]
Abstract
A mathematical model of the interaction of propafenone with cardiac sodium channels is based on experimental data that demonstrate use-dependent effects of the drug. The Clancy-Rudy model is applied to describe Na-channels in absence of the drug. The values of rate constants of the drug-receptor reaction are fitted to experimental data by iterative computer simulations using a genetic algorithm. The model suggests the following interpretation of available experimental results: First, drug molecules have access to the binding sites predominantly in the inactivated states. Secondly, the biphasic development of the block during depolarisation is consistent with a rapid increase due to drug binding in the fast inactivated state (rate constants k(on) = 645 micromol(-1) l s(-1), k(off) = 16.21 s(-1)) and a slow increase due to binding in the intermediate inactivated state (rate constants approximately 100-fold lower), followed by transition to the drug-occupied slow inactivated state (rate constants 0.784 and 0.921 s(-1)). Thirdly, the observed biphasic time course of recovery of I(Na) from block following restoration of the resting voltage results from simultaneous relief of block from the channels residing in the intermediate and slow inactivated states. Fourthly, the accumulation of blocked channels in the slow inactivated state is responsible for the observed use-dependent effects. Fifthly, when incorporated into a quantitative description of the electrical activity of a ventricular cell, the model predicts that propafenone (0.2 micromol l(-1)) effectively suppresses premature excitations, leaving the regular action potentials nearly unaffected.
Collapse
Affiliation(s)
- M Pásek
- Institute of Thermomechanics, Czech Academy of Science, Branch Brno, Czech Republic.
| | | |
Collapse
|
41
|
Wang GK, Russell C, Wang SY. State-dependent block of voltage-gated Na+ channels by amitriptyline via the local anesthetic receptor and its implication for neuropathic pain. Pain 2004; 110:166-74. [PMID: 15275764 DOI: 10.1016/j.pain.2004.03.018] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2003] [Revised: 02/10/2004] [Accepted: 03/09/2004] [Indexed: 11/28/2022]
Abstract
Amitriptyline is a tricyclic antidepressant, which also alleviates various pain syndromes at its therapeutic plasma concentration (0.36-0.90 microM). Accumulated evidence suggests that such efficacy may be due to block of voltage-gated Na(+) channels. The Na(+) channel alpha-subunit protein consists of four homologous domains (D1-D4), each with six transmembrane segments (S1-S6). The aims of this study were to locate the amitriptyline receptor in the Na(+) channel alpha-subunit and to compare the amitriptyline affinity in open, inactivated, and resting states of the Na(+) channel. Wild-type and mutant rat skeletal muscle alpha-subunit Na(+) channels were expressed in human embryonic kidney cells and assayed under whole-cell voltage clamp conditions. Our results indicate that the amitriptyline receptor overlaps with the local anesthetic receptor to a great extent in Na(+) channels. Residues N434 (at D1-S6), L1280 (D3-S6), and F1579 (D4-S6) may jointly form parts of the amitriptyline/local anesthetic receptor, with residue L1280 being most critical for amitriptyline binding. Open-channel block by amitriptyline was assessed in inactivation-deficient Na(+) channels and compared with the resting- and inactivated-channel block in wild-type channels. The open-channel block by amitriptyline has the highest affinity, with a 50% inhibitory concentration (IC(50)) of 0.26 microM. The inactivated-channel block by amitriptyline had a weaker affinity (0.51 microM), whereas the resting-channel displayed the weakest affinity (33 microM). We hypothesize that selective block of both persistent late openings and the inactivated state of neuronal Na(+) channel isoforms by amitriptyline also occurs at its therapeutic concentration and likely contributes to its efficacy in pain syndromes.
Collapse
Affiliation(s)
- Ging Kuo Wang
- Department of Anesthesia, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | | | | |
Collapse
|
42
|
Zarrindast MR, Baghdadi B, Sahebgharani M. Potentiation of imipramine-induced antinociception by nicotine in the formalin test. Eur Neuropsychopharmacol 2004; 14:71-6. [PMID: 14659990 DOI: 10.1016/s0924-977x(03)00066-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In this study, the effect of cholinergic agents on imipramine antinociception in mice, in the formalin test, has been investigated. Intraperitoneal (i.p.) administration of different doses of imipramine (2.5, 5, 10, 20 and 30 mg/kg) or nicotine (0.25, 0.5, 0.75 and 1 mg/kg) induced a dose dependent antinociception in both the first and second phases of the formalin test in mice. The combination of imipramine with doses of 0.5 and 0.75 mg/kg of nicotine showed a potentiated response, in both phases of the test. However, neither hexamethonium (5 and 10 mg/kg), atropine (0.25 mg/kg) or mecamylamine (0.25 mg/kg) altered the antinociception induced by imipramine. It is concluded that nicotinic receptor activation but not the cholinergic muscarinic mechanism is involved in the imipramine-induced antinociception.
Collapse
Affiliation(s)
- Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784 Tehran, Iran.
| | | | | |
Collapse
|
43
|
Abstract
Genetic variability has recently been implicated in the development of familial epilepsy syndromes and in heterogeneous responses of epilepsy patients to drug treatment. Mutations in distinct proteins have been shown to underlie the development of epilepsy, increase propensity for drug resistance, and alter drug metabolism. Improved understanding of how individual genetic variability may alter the efficacy of pharmacological therapeutic interventions is an important and timely goal. The investigation of relationships between genotype and patient responses to drug treatment is termed pharmacogenomics.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Pharmacology, Columbia University, College of Physicians and Surgeons, 630 W. 168th Street, New York, NY 10032, USA.
| | | |
Collapse
|
44
|
De Luca A, Talon S, De Bellis M, Desaphy JF, Lentini G, Corbo F, Scilimati A, Franchini C, Tortorella V, Camerino DC. Optimal requirements for high affinity and use-dependent block of skeletal muscle sodium channel by N-benzyl analogs of tocainide-like compounds. Mol Pharmacol 2003; 64:932-45. [PMID: 14500750 DOI: 10.1124/mol.64.4.932] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Newly synthesized tocainide analogs were tested for their state-dependent affinity and use-dependent behavior on sodium currents (INa) of adult skeletal muscle fibers by means of the Vaseline-gap voltage clamp method. The drugs had the pharmacophore amino group constrained in position alpha [N-(2,6-dimethylphenyl)pyrrolidine-2-carboxamide (To5)] or beta [N-(2,6-dimethylphenyl)pyrrolidine-3-carboxamide (To9)] in a proline-like cycle and/or linked to a lipophilic benzyl moiety as in N-benzyl-tocainide (Benzyl-Toc), 1-benzyl-To5 (Benzyl-To5), and 1-benzyl-To9 (Benzyl-To9). INa were elicited with pulses to -20 mV from different holding potentials (-140, -100, and -70 mV) and stimulation frequencies (2 and 10 Hz). All compounds were voltage-dependent and use-dependent channel blockers. The presence of a proline-like cycle increased the potency; i.e., To5 was 3- and 10-fold more effective than Toc in blocking INa at the holding potential of -140 and -70 mV, respectively. The benzyl group on the amine further enhanced drug effectiveness with the following scale: Benzyl-To9 >/= Benzyl-Toc > Benzyl-To5. At a holding potential of -100 mV and 10-Hz stimulation, Benzyl-To9 blocked INa with a half-maximal concentration of 0.5 microM, being 60 and 400 times more potent than To9 and Toc, respectively. The similar effectiveness of Benzyl-Toc and Benzyl-To9 was paralleled by a similar spatial arrangement by equilibrium geometry modeling. In addition, the latter had a higher pKa value that probably contributed to a slow kinetic during its high use-dependent behavior. Benzyl-To5 had its lowest energy level at a more folded conformation that justifies the less favorable profile among the N-benzylated analogs. The new compounds are the most potent tocainide-like sodium channel blockers so far described and have high therapeutic potentials.
Collapse
Affiliation(s)
- Annamaria De Luca
- Unità di Farmacologia, Dipartimento Farmacobiologico, Facoltà di Farmacia, University di Bari, Via Orabona 4, Campus 70121, Bari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|